EP3922722A1 - Composés antisens sélectifs et leurs utilisations - Google Patents

Composés antisens sélectifs et leurs utilisations Download PDF

Info

Publication number
EP3922722A1
EP3922722A1 EP21161967.1A EP21161967A EP3922722A1 EP 3922722 A1 EP3922722 A1 EP 3922722A1 EP 21161967 A EP21161967 A EP 21161967A EP 3922722 A1 EP3922722 A1 EP 3922722A1
Authority
EP
European Patent Office
Prior art keywords
nucleoside
certain embodiments
sugar moiety
modified
oligomeric compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP21161967.1A
Other languages
German (de)
English (en)
Other versions
EP3922722B1 (fr
Inventor
Punit P. Seth
Michael OESTERGAARD
Eric E Swayze
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ionis Pharmaceuticals Inc
Original Assignee
Ionis Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46717938&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP3922722(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Ionis Pharmaceuticals Inc filed Critical Ionis Pharmaceuticals Inc
Priority to EP23181830.3A priority Critical patent/EP4269584A3/fr
Publication of EP3922722A1 publication Critical patent/EP3922722A1/fr
Application granted granted Critical
Publication of EP3922722B1 publication Critical patent/EP3922722B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/312Phosphonates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/312Phosphonates
    • C12N2310/3125Methylphosphonates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/314Phosphoramidates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/316Phosphonothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/34Allele or polymorphism specific uses

Definitions

  • the present invention pertains generally to chemically-modified oligonucleotides for use in research, diagnostics, and/or therapeutics.
  • Antisense compounds have been used to modulate target nucleic acids. Antisense compounds comprising a variety of chemical modifications and motifs have been reported. In certain instances, such compounds are useful as research tools, diagnostic reagents, and as therapeutic agents. In certain instances antisense compounds have been shown to modulate protein expression by binding to a target messenger RNA (mRNA) encoding the protein. In certain instances, such binding of an antisense compound to its target mRNA results in cleavage of the mRNA. Antisense compounds that modulate processing of a pre-mRNA have also been reported. Such antisense compounds alter splicing, interfere with polyadenlyation or prevent formation of the 5'-cap of a pre-mRNA.
  • mRNA target messenger RNA
  • the present invention provides oligomeric compounds comprising oligonucleotides.
  • such oligonucleotides comprise a region having a gapmer motif.
  • such oligonucleotides consist of a region having a gapmer motif.
  • oligomeric compounds including oligonucleotides described herein are capable of modulating expression of a target RNA.
  • the target RNA is associated with a disease or disorder, or encodes a protein that is associated with a disease or disorder.
  • the oligomeric compounds or oligonucleotides provided herein modulate the expression of function of such RNA to alleviate one or more symptom of the disease or disorder.
  • oligomeric compounds including oligonucleotides describe herein are useful in vitro. In certain embodiments such oligomeric compounds are used in diagnostics and/or for target validation experiments.
  • nucleoside means a compound comprising a nucleobase moiety and a sugar moiety. Nucleosides include, but are not limited to, naturally occurring nucleosides (as found in DNA and RNA) and modified nucleosides. Nucleosides may be linked to a phosphate moiety.
  • chemical modification means a chemical difference in a compound when compared to a naturally occurring counterpart.
  • Chemical modifications of oligonucleotides include nucleoside modifications (including sugar moiety modifications and nucleobase modifications) and internucleoside linkage modifications. In reference to an oligonucleotide, chemical modification does not include differences only in nucleobase sequence.
  • furanosyl means a structure comprising a 5-membered ring comprising four carbon atoms and one oxygen atom.
  • sugar moiety means a naturally occurring sugar moiety or a modified sugar moiety of a nucleoside.
  • modified sugar moiety means a substituted sugar moiety or a sugar surrogate.
  • substituted sugar moiety means a furanosyl that is not a naturally occurring sugar moiety.
  • Substituted sugar moieties include, but are not limited to furanosyls comprising substituents at the 2'-position, the 3'-position, the 5'-position and/or the 4'-position.
  • Certain substituted sugar moieties are bicyclic sugar moieties.
  • 2'-substituted sugar moiety means a furanosyl comprising a substituent at the 2'-position other than H or OH. Unless otherwise indicated, a 2'-substituted sugar moiety is not a bicyclic sugar moiety (i.e., the 2'-substituent of a 2'-substituted sugar moiety does not form a bridge to another atom of the furanosyl ring.
  • MOE means -OCH 2 CH 2 OCH 3 .
  • 2'-F nucleoside refers to a nucleoside comprising a sugar comprising fluoroine at the 2' position. Unless otherwise indicated, the fluorine in a 2'-F nucleoside is in the ribo position (replacing the OH of a natural ribose).
  • 2'-(ara)-F refers to a 2'-F substituted nucleoside, wherein the fluoro group is in the arabino position.
  • sucrose surrogate means a structure that does not comprise a furanosyl and that is capable of replacing the naturally occurring sugar moiety of a nucleoside, such that the resulting nucleoside sub-units are capable of linking together and/or linking to other nucleosides to form an oligomeric compound which is capable of hybridizing to a complementary oligomeric compound.
  • Such structures include rings comprising a different number of atoms than furanosyl (e.g., 4, 6, or 7-membered rings); replacement of the oxygen of a furanosyl with a non-oxygen atom (e.g., carbon, sulfur, or nitrogen); or both a change in the number of atoms and a replacement of the oxygen.
  • Such structures may also comprise substitutions corresponding to those described for substituted sugar moieties (e.g., 6-membered carbocyclic bicyclic sugar surrogates optionally comprising additional substituents).
  • Sugar surrogates also include more complex sugar replacements (e.g., the non-ring systems of peptide nucleic acid).
  • Sugar surrogates include without limitation morpholinos, cyclohexenyls and cyclohexitols.
  • bicyclic sugar moiety means a modified sugar moiety comprising a 4 to 7 membered ring (including but not limited to a furanosyl) comprising a bridge connecting two atoms of the 4 to 7 membered ring to form a second ring, resulting in a bicyclic structure.
  • the 4 to 7 membered ring is a sugar ring.
  • the 4 to 7 membered ring is a furanosyl.
  • the bridge connects the 2'-carbon and the 4'-carbon of the furanosyl.
  • nucleotide means a nucleoside further comprising a phosphate linking group.
  • linked nucleosides may or may not be linked by phosphate linkages and thus includes, but is not limited to “linked nucleotides.”
  • linked nucleosides are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked).
  • nucleobase means a group of atoms that can be linked to a sugar moiety to create a nucleoside that is capable of incorporation into an oligonucleotide, and wherein the group of atoms is capable of bonding with a complementary naturally occurring nucleobase of another oligonucleotide or nucleic acid. Nucleobases may be naturally occurring or may be modified.
  • unmodified nucleobase or “naturally occurring nucleobase” means the naturally occurring heterocyclic nucleobases of RNA or DNA: the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) (including 5-methyl C), and uracil (U).
  • modified nucleobase means any nucleobase that is not a naturally occurring nucleobase.
  • modified nucleoside means a nucleoside comprising at least one chemical modification compared to naturally occurring RNA or DNA nucleosides. Modified nucleosides comprise a modified sugar moiety and/or a modified nucleobase.
  • bicyclic nucleoside or "BNA” means a nucleoside comprising a bicyclic sugar moiety.
  • constrained ethyl nucleoside or “cEt” means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH(CH 3 )-O-2'bridge.
  • locked nucleic acid nucleoside or "LNA” means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH 2 -O-2'bridge.
  • 2'-substituted nucleoside means a nucleoside comprising a substituent at the 2'-position other than H or OH. Unless otherwise indicated, a 2'-substituted nucleoside is not a bicyclic nucleoside.
  • 2'-deoxynucleoside means a nucleoside comprising 2'-H furanosyl sugar moiety, as found in naturally occurring deoxyribonucleosides (DNA).
  • a 2'-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (e.g., uracil).
  • RNA-like nucleoside means a modified nucleoside that adopts a northern configuration and functions like RNA when incorporated into an oligonucleotide.
  • RNA-like nucleosides include, but are not limited to 3'-endo furanosyl nucleosides and RNA surrogates.
  • 3'-endo-furanosyl nucleoside means an RNA-like nucleoside that comprises a substituted sugar moiety that has a 3'-endo conformation.
  • 3'-endo-furanosyl nucleosides include, but are not limitied to: 2'-MOE, 2'-F, 2'-OMe, LNA, ENA, and cEt nucleosides.
  • RNA-surrogate nucleoside means an RNA-like nucleoside that does not comprise a furanosyl. RNA-surrogate nucleosides include, but are not limited to hexitols and cyclopentanes.
  • oligonucleotide means a compound comprising a plurality of linked nucleosides.
  • an oligonucleotide comprises one or more unmodified ribonucleosides (RNA) and/or unmodified deoxyribonucleosides (DNA) and/or one or more modified nucleosides.
  • oligonucleoside means an oligonucleotide in which none of the internucleoside linkages contains a phosphorus atom.
  • oligonucleotides include oligonucleosides.
  • modified oligonucleotide means an oligonucleotide comprising at least one modified nucleoside and/or at least one modified internucleoside linkage.
  • nucleoside linkage means a covalent linkage between adjacent nucleosides in an oligonucleotide.
  • naturally occurring internucleoside linkage means a 3' to 5' phosphodiester linkage.
  • modified internucleoside linkage means any internucleoside linkage other than a naturally occurring internucleoside linkage.
  • oligomeric compound means a polymeric structure comprising two or more substructures.
  • an oligomeric compound comprises an oligonucleotide.
  • an oligomeric compound comprises one or more conjugate groups and/or terminal groups.
  • an oligomeric compound consists of an oligonucleotide.
  • terminal group means one or more atom attached to either, or both, the 3' end or the 5' end of an oligonucleotide. In certain embodiments a terminal group is a conjugate group. In certain embodiments, a terminal group comprises one or more terminal group nucleosides.
  • conjugate means an atom or group of atoms bound to an oligonucleotide or oligomeric compound.
  • conjugate groups modify one or more properties of the compound to which they are attached, including, but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and/or clearance properties.
  • conjugate linking group means any atom or group of atoms used to attach a conjugate to an oligonucleotide or oligomeric compound.
  • antisense compound means a compound comprising or consisting of an oligonucleotide at least a portion of which is complementary to a target nucleic acid to which it is capable of hybridizing, resulting in at least one antisense activity.
  • antisense activity means any detectable and/or measurable change attributable to the hybridization of an antisense compound to its target nucleic acid.
  • detecting or “measuring” means that a test or assay for detecting or measuring is performed. Such detection and/or measuring may result in a value of zero. Thus, if a test for detection or measuring results in a finding of no activity (activity of zero), the step of detecting or measuring the activity has nevertheless been performed.
  • detectable and/or measureable activity means a measurable activity that is not zero.
  • essentially unchanged means little or no change in a particular parameter, particularly relative to another parameter which changes much more.
  • a parameter is essentially unchanged when it changes less than 5%.
  • a parameter is essentially unchanged if it changes less than two-fold while another parameter changes at least ten-fold.
  • an antisense activity is a change in the amount of a target nucleic acid.
  • the amount of a non-target nucleic acid is essentially unchanged if it changes much less than the target nucleic acid does, but the change need not be zero.
  • expression means the process by which a gene ultimately results in a protein.
  • Expression includes, but is not limited to, transcription, post-transcriptional modification (e.g., splicing, polyadenlyation, addition of 5'-cap), and translation.
  • target nucleic acid means a nucleic acid molecule to which an antisense compound is intended to hybridize.
  • non-target nucleic acid means a nucleic acid molecule to which hybridization of an antisense compound is not intended or desired.
  • antisense compounds do hybridize to a non-target, due to homology between the target (intended) and non-target (un-intended).
  • mRNA means an RNA molecule that encodes a protein.
  • pre-mRNA means an RNA transcript that has not been fully processed into mRNA. Pre-RNA includes one or more intron.
  • object RNA means an RNA molecule other than a target RNA, the amount, activity, splicing, and/or function of which is modulated, either directly or indirectly, by a target nucleic acid.
  • a target nucleic acid modulates splicing of an object RNA.
  • an antisense compound modulates the amount or activity of the target nucleic acid, resulting in a change in the splicing of an object RNA and ultimately resulting in a change in the activity or function of the object RNA.
  • microRNA means a naturally occurring, small, non-coding RNA that represses gene expression of at least one mRNA.
  • a microRNA represses gene expression by binding to a target site within a 3' untranslated region of an mRNA.
  • a microRNA has a nucleobase sequence as set forth in miRBase, a database of published microRNA sequences found at http://microrna.sanger.ac.uk/sequences/.
  • a microRNA has a nucleobase sequence as set forth in miRBase version 12.0 released September 2008, which is herein incorporated by reference in its entirety.
  • differentiating nucleobase means a nucleobase that differs between two nucleic acids.
  • a target region of a target nucleic acid differs by 1-4 nucleobases from a non-target nucleic acid. Each of those differences is refered to as a differentiating nucleobase.
  • a differentiating nucleobase is a single-nucleotide polymorphism.
  • target-selective nucleoside means a nucleoside of an antisense compound that corresponds to a differentiating nucleobase of a target nucleic acid.
  • allelic pair means one of a pair of copies of a gene existing at a particular locus or marker on a specific chromosome, or one member of a pair of nucleobases existing at a particular locus or marker on a specific chromosome, or one member of a pair of nucleobase sequences existing at a particular locus or marker on a specific chromosome.
  • each allelic pair will normally occupy corresponding positions (loci) on a pair of homologous chromosomes, one inherited from the mother and one inherited from the father.
  • the organism or cell is said to be “homozygous” for that allele; if they differ, the organism or cell is said to be “heterozygous” for that allele.
  • Wild-type allele refers to the genotype typically not associated with disease or dysfunction of the gene product.
  • Melt allele refers to the genotype associated with disease or dysfunction of the gene product.
  • allelic variant means a particular identity of an allele, where more than one identity occurs.
  • an allelic variant may refer to either the mutant allele or the wild-type allele.
  • single nucleotide polymorphism or "SNP” means a single nucleotide variation between the genomes of individuals of the same species.
  • a SNP may be a single nucleotide deletion or insertion.
  • SNPs occur relatively frequently in genomes and thus contribute to genetic diversity. The location of a SNP is generally flanked by highly conserved sequences. An individual may be homozygous or heterozygous for an allele at each SNP site.
  • single nucleotide polymorphism site or “SNP site” refers to the nucleotides surrounding a SNP contained in a target nucleic acid to which an antisense compound is targeted.
  • targeting means the association of an antisense compound to a particular target nucleic acid molecule or a particular region of a target nucleic acid molecule.
  • An antisense compound targets a target nucleic acid if it is sufficiently complementary to the target nucleic acid to allow hybridization under physiological conditions.
  • nucleobase complementarity or “complementarity” when in reference to nucleobases means a nucleobase that is capable of base pairing with another nucleobase.
  • adenine (A) is complementary to thymine (T).
  • adenine (A) is complementary to uracil (U).
  • complementary nucleobase means a nucleobase of an antisense compound that is capable of base pairing with a nucleobase of its target nucleic acid.
  • nucleobases at a certain position of an antisense compound are capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid
  • the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be complementary at that nucleobase pair.
  • Nucleobases comprising certain modifications may maintain the ability to pair with a counterpart nucleobase and thus, are still capable of nucleobase complementarity.
  • non-complementary in reference to nucleobases means a pair of nucleobases that do not form hydrogen bonds with one another.
  • complementary in reference to oligomeric compounds (e.g., linked nucleosides, oligonucleotides, or nucleic acids) means the capacity of such oligomeric compounds or regions thereof to hybridize to another oligomeric compound or region thereof through nucleobase complementarity under stringent conditions.
  • Complementary oligomeric compounds need not have nucleobase complementarity at each nucleoside. Rather, some mismatches are tolerated.
  • complementary oligomeric compounds or regions are complementary at 70% of the nucleobases (70% complementary).
  • complementary oligomeric compounds or regions are 80% complementary.
  • complementary oligomeric compounds or regions are 90% complementary.
  • complementary oligomeric compounds or regions are 95% complementary.
  • complementary oligomeric compounds or regions are 100% complementary.
  • mismatch means a nucleobase of a first oligomeric compound that is not capable of pairing with a nucleobase at a corresponding position of a second oligomeric compound, when the first and second oligomeric compound are aligned.
  • first and second oligomeric compounds may be oligonucleotides.
  • hybridization means the pairing of complementary oligomeric compounds (e.g., an antisense compound and its target nucleic acid). While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleobases.
  • oligomeric compound specifically hybridizes to more than one target site.
  • oligonucleotide or portion thereof means that each nucleobase of the oligonucleotide or portion thereof is capable of pairing with a nucleobase of a complementary nucleic acid or contiguous portion thereof.
  • a fully complementary region comprises no mismatches or unhybridized nucleobases in either strand.
  • percent complementarity means the percentage of nucleobases of an oligomeric compound that are complementary to an equal-length portion of a target nucleic acid. Percent complementarity is calculated by dividing the number of nucleobases of the oligomeric compound that are complementary to nucleobases at corresponding positions in the target nucleic acid by the total length of the oligomeric compound.
  • percent identity means the number of nucleobases in a first nucleic acid that are the same type (independent of chemical modification) as nucleobases at corresponding positions in a second nucleic acid, divided by the total number of nucleobases in the first nucleic acid.
  • modulation means a change of amount or quality of a molecule, function, or activity when compared to the amount or quality of a molecule, function, or activity prior to modulation.
  • modulation includes the change, either an increase (stimulation or induction) or a decrease (inhibition or reduction) in gene expression.
  • modulation of expression can include a change in splice site selection of pre-mRNA processing, resulting in a change in the absolute or relative amount of a particular splice-variant compared to the amount in the absence of modulation.
  • modification motif means a pattern of chemical modifications in an oligomeric compound or a region thereof. Motifs may be defined by modifications at certain nucleosides and/or at certain linking groups of an oligomeric compound.
  • nucleoside motif means a pattern of nucleoside modifications in an oligomeric compound or a region thereof.
  • the linkages of such an oligomeric compound may be modified or unmodified.
  • motifs herein describing only nucleosides are intended to be nucleoside motifs. Thus, in such instances, the linkages are not limited.
  • sugar motif means a pattern of sugar modifications in an oligomeric compound or a region thereof.
  • linkage motif means a pattern of linkage modifications in an oligomeric compound or region thereof.
  • the nucleosides of such an oligomeric compound may be modified or unmodified.
  • motifs herein describing only linkages are intended to be linkage motifs. Thus, in such instances, the nucleosides are not limited.
  • nucleobase modification motif means a pattern of modifications to nucleobases along an oligonucleotide. Unless otherwise indicated, a nucleobase modification motif is independent of the nucleobase sequence.
  • sequence motif means a pattern of nucleobases arranged along an oligonucleotide or portion thereof. Unless otherwise indicated, a sequence motif is independent of chemical modifications and thus may have any combination of chemical modifications, including no chemical modifications.
  • nucleoside having a modification of a first type may be an unmodified nucleoside.
  • telomeres As used herein, “differently modified” mean chemical modifications or chemical substituents that are different from one another, including absence of modifications. Thus, for example, a MOE nucleoside and an unmodified DNA nucleoside are “differently modified,” even though the DNA nucleoside is unmodified. Likewise, DNA and RNA are “differently modified,” even though both are naturally-occurring unmodified nucleosides. Nucleosides that are the same but for comprising different nucleobases are not differently modified.
  • nucleoside comprising a 2'-OMe modified sugar and an unmodified adenine nucleobase and a nucleoside comprising a 2'-OMe modified sugar and an unmodified thymine nucleobase are not differently modified.
  • the same type of modifications refers to modifications that are the same as one another, including absence of modifications.
  • two unmodified DNA nucleoside have “the same type of modification,” even though the DNA nucleoside is unmodified.
  • Such nucleosides having the same type modification may comprise different nucleobases.
  • pharmaceutically acceptable carrier or diluent means any substance suitable for use in administering to an animal.
  • a pharmaceutically acceptable carrier or diluent is sterile saline.
  • such sterile saline is pharmaceutical grade saline.
  • substituted nucleoside and “substituent group,” means an atom or group that replaces the atom or group of a named parent compound.
  • a substituent of a modified nucleoside is any atom or group that differs from the atom or group found in a naturally occurring nucleoside (e.g., a modified 2'-substuent is any atom or group at the 2'-position of a nucleoside other than H or OH).
  • Substituent groups can be protected or unprotected.
  • compounds of the present invention have substituents at one or at more than one position of the parent compound. Substituents may also be further substituted with other substituent groups and may be attached directly or via a linking group such as an alkyl or hydrocarbyl group to a parent compound.
  • substituted in reference to a chemical functional group means an atom or group of atoms differs from the atom or a group of atoms normally present in the named functional group.
  • a substituent replaces a hydrogen atom of the functional group (e.g., in certain embodiments, the substituent of a substituted methyl group is an atom or group other than hydrogen which replaces one of the hydrogen atoms of an unsubstituted methyl group).
  • R aa , R bb and R cc is, independently, H, an optionally linked chemical functional group or a further substituent group with a preferred list including without limitation, alkyl
  • alkyl means a saturated straight or branched hydrocarbon radical containing up to twenty four carbon atoms.
  • alkyl groups include without limitation, methyl, ethyl, propyl, butyl, isopropyl, n-hexyl, octyl, decyl, dodecyl and the like.
  • Alkyl groups typically include from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms (C 1 -C 12 alkyl) with from 1 to about 6 carbon atoms being more preferred.
  • alkenyl means a straight or branched hydrocarbon chain radical containing up to twenty four carbon atoms and having at least one carbon-carbon double bond.
  • alkenyl groups include without limitation, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, dienes such as 1,3-butadiene and the like.
  • Alkenyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred.
  • Alkenyl groups as used herein may optionally include one or more further substituent groups.
  • alkynyl means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms and having at least one carbon-carbon triple bond.
  • alkynyl groups include, without limitation, ethynyl, 1-propynyl, 1-butynyl, and the like.
  • Alkynyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred.
  • Alkynyl groups as used herein may optionally include one or more further substituent groups.
  • acyl means a radical formed by removal of a hydroxyl group from an organic acid and has the general Formula -C(O)-X where X is typically aliphatic, alicyclic or aromatic. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates, aliphatic phosphates and the like. Acyl groups as used herein may optionally include further substituent groups.
  • alicyclic means a cyclic ring system wherein the ring is aliphatic.
  • the ring system can comprise one or more rings wherein at least one ring is aliphatic.
  • Preferred alicyclics include rings having from about 5 to about 9 carbon atoms in the ring.
  • Alicyclic as used herein may optionally include further substituent groups.
  • aliphatic means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms wherein the saturation between any two carbon atoms is a single, double or triple bond.
  • An aliphatic group preferably contains from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred.
  • the straight or branched chain of an aliphatic group may be interrupted with one or more heteroatoms that include nitrogen, oxygen, sulfur and phosphorus.
  • Such aliphatic groups interrupted by heteroatoms include without limitation, polyalkoxys, such as polyalkylene glycols, polyamines, and polyimines. Aliphatic groups as used herein may optionally include further substituent groups.
  • alkoxy means a radical formed between an alkyl group and an oxygen atom wherein the oxygen atom is used to attach the alkoxy group to a parent molecule.
  • alkoxy groups include without limitation, methoxy, ethoxy, propoxy, isopropoxy, n -butoxy, sec-butoxy, tert -butoxy, n-pentoxy, neopentoxy, n-hexoxy and the like.
  • Alkoxy groups as used herein may optionally include further substituent groups.
  • aminoalkyl means an amino substituted C 1 -C 12 alkyl radical.
  • the alkyl portion of the radical forms a covalent bond with a parent molecule.
  • the amino group can be located at any position and the aminoalkyl group can be substituted with a further substituent group at the alkyl and/or amino portions.
  • aralkyl and arylalkyl mean an aromatic group that is covalently linked to a C 1 -C 12 alkyl radical.
  • the alkyl radical portion of the resulting aralkyl (or arylalkyl) group forms a covalent bond with a parent molecule. Examples include without limitation, benzyl, phenethyl and the like.
  • Aralkyl groups as used herein may optionally include further substituent groups attached to the alkyl, the aryl or both groups that form the radical group.
  • aryl and aromatic mean a mono- or polycyclic carbocyclic ring system radicals having one or more aromatic rings.
  • aryl groups include without limitation, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like.
  • Preferred aryl ring systems have from about 5 to about 20 carbon atoms in one or more rings.
  • Aryl groups as used herein may optionally include further substituent groups.
  • heteroaryl and “heteroaromatic,” mean a radical comprising a mono- or polycyclic aromatic ring, ring system or fused ring system wherein at least one of the rings is aromatic and includes one or more heteroatoms. Heteroaryl is also meant to include fused ring systems including systems where one or more of the fused rings contain no heteroatoms. Heteroaryl groups typically include one ring atom selected from sulfur, nitrogen or oxygen.
  • heteroaryl groups include without limitation, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl and the like.
  • Heteroaryl radicals can be attached to a parent molecule directly or through a linking moiety such as an aliphatic group or hetero atom.
  • Heteroaryl groups as used herein may optionally include further substituent groups.
  • the present invention provides oligomeric compounds.
  • such oligomeric compounds comprise oligonucleotides optionally comprising one or more conjugate and/or terminal groups.
  • an oligomeric compound consists of an oligonucleotide.
  • oligonucleotides comprise one or more chemical modifications. Such chemical modifications include modifications of one or more nucleoside (including modifications to the sugar moiety and/or the nucleobase) and/or modifications to one or more internucleoside linkage.
  • oligomeric compounds comprising or consisting of oligonuleotides comprising at least one modified nucleoside.
  • modified nucleosides comprise a modified sugar moeity, a modified nucleobase, or both a modifed sugar moiety and a modified nucleobase.
  • compounds of the invention comprise one or more modifed nucleosides comprising a modifed sugar moiety.
  • Such compounds comprising one or more sugar-modified nucleosides may have desirable properties, such as enhanced nuclease stability or increased binding affinity with a target nucleic acid relative to an oligonucleotide comprising only nucleosides comprising naturally occurring sugar moieties.
  • modified sugar moieties are substitued sugar moieties.
  • modified sugar moieties are sugar surrogates.
  • Such sugar surogates may comprise one or more substitutions corresponding to those of substituted sugar moieties.
  • modified sugar moieties are substituted sugar moieties comprising one or more non-bridging sugar substituent, including but not limited to substituents at the 2' and/or 5' positions.
  • sugar substituents suitable for the 2'-position include, but are not limited to: 2'-F, 2'-OCH 3 ("OMe” or "O-methyl"), and 2'-O(CH 2 ) 2 OCH 3 (“MOE").
  • sugar substituents at the 5'-position include, but are not limited to:, 5'-methyl (R or S); 5'-vinyl, and 5'-methoxy.
  • substituted sugars comprise more than one non-bridging sugar substituent, for example, 2'-F-5'-methyl sugar moieties (see,e.g., PCT International Application WO 2008/101157 , for additional 5', 2'-bis substituted sugar moieties and nucleosides).
  • Nucleosides comprising 2'-substituted sugar moieties are referred to as 2'-substituted nucleosides.
  • These 2'-substituent groups can be further substituted with one or more substituent groups independently selected from hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO 2 ), thiol, thioalkoxy (S-alkyl), halogen, alkyl, aryl, alkenyl and alkynyl.
  • a 2'- substituted nucleoside comprises a sugar moiety comprising a 2'-substituent group selected from F, OCF 3 , O-CH 3 , OCH 2 CH 2 OCH 3 , O(CH 2 ) 2 SCH 3 , O-(CH 2 ) 2 -O-N(CH 3 ) 2 , -O(CH 2 ) 2 O(CH 2 ) 2 N(CH 3 ) 2 , and O-CH 2
  • a 2'- substituted nucleoside comprises a sugar moiety comprising a 2'-substituent group selected from F, O-CH 3 , and OCH 2 CH 2 OCH 3 .
  • Certain modifed sugar moieties comprise a bridging sugar substituent that forms a second ring resulting in a bicyclic sugar moiety.
  • the bicyclic sugar moiety comprises a bridge between the 4' and the 2' furanose ring atoms.
  • Examples of such 4' to 2' sugar substituents include, but are not limited to: -[C(R a )(R b )] n -, -[C(R a )(R b )] n -O-, -C(R a R b )-N(R)-O- or, -C(R a R b )-O-N(R)-; 4'-CH 2 -2', 4'-(CH 2 ) 2 -2', 4'-(CH 2 ) 3 -2',.
  • Bicyclic nucleosides include, but are not limited to, (A) ⁇ -L-Methyleneoxy (4'-CH 2 -O-2') BNA, (B) ⁇ -D-Methyleneoxy (4'-CH 2 -O-2') BNA (also referred to as locked nucleic acid or LNA), (C) Ethyleneoxy (4'-(CH 2 ) 2 -O-2') BNA, (D) Aminooxy (4'-CH 2 -O-N(R)-2') BNA, (E) Oxyamino (4'-CH 2 -N(R)-O-2') BNA, (F) Methyl(methyleneoxy) (4'-CH(CH 3 )-O-2') BNA (also referred to as constrained ethyl or cEt), (G) methylene-thio (4'-CH 2 -S
  • bicyclic sugar moieties and nucleosides incorporating such bicyclic sugar moieties are further defined by isomeric configuration.
  • a nucleoside comprising a 4'-2' methylene-oxy bridge may be in the ⁇ -L configuration or in the ⁇ -D configuration.
  • ⁇ -L-methyleneoxy (4'-CH 2 -O-2') bicyclic nucleosides have been incorporated into antisense oligonucleotides that showed antisense activity ( Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372 ).
  • substituted sugar moieties comprise one or more non-bridging sugar substituent and one or more bridging sugar substituent (e.g., 5'-substituted and 4'-2' bridged sugars).
  • bridging sugar substituent e.g., 5'-substituted and 4'-2' bridged sugars.
  • modified sugar moieties are sugar surrogates.
  • the oxygen atom of the naturally occuring sugar is substituted, e.g., with a sulfer, carbon or nitrogen atom.
  • such modified sugar moiety also comprises bridging and/or non-bridging substituents as described above.
  • certain sugar surogates comprise a 4'-sulfer atom and a substitution at the 2'-position ( see,e.g., published U.S. Patent Application US2005/0130923, published on June 16, 2005 ) and/or the 5' position.
  • carbocyclic bicyclic nucleosides having a 4'-2' bridge have been described (see, e.g., Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., J. Org. Chem., 2006, 71, 7731-7740 ).
  • sugar surrogates comprise rings having other than 5-atoms.
  • a sugar surrogate comprises a six-membered tetrahydropyran.
  • Such tetrahydropyrans may be further modified or substituted.
  • Nucleosides comprising such modified tetrahydropyrans include, but are not limited to, hexitol nucleic acid (HNA), anitol nucleic acid (ANA), manitol nucleic acid (MNA) ( see Leumann, CJ. Bioorg. & Med. Chem. (2002) 10:841-854 ), fluoro HNA (F-HNA), and those compounds having Formula VII: wherein independently for each of said at least one tetrahydropyran nucleoside analog of Formula VII:
  • the modified THP nucleosides of Formula VII are provided wherein q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 are each H. In certain embodiments, at least one of q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 is other than H. In certain embodiments, at least one of q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 is methyl. In certain embodiments, THP nucleosides of Formula VII are provided wherein one of R 1 and R 2 is F. In certain embodiments, R 1 is fluoro and R 2 is H, R 1 is methoxy and R 2 is H, and R 1 is methoxyethoxy and R 2 is H.
  • Patent Application US2005-0130923, published on June 16, 2005 ) or alternatively 5'-substitution of a bicyclic nucleic acid see PCT International Application WO 2007/134181 , published on 11/22/07 wherein a 4'-CH 2 -O-2' bicyclic nucleoside is further substituted at the 5' position with a 5'-methyl or a 5'-vinyl group).
  • PCT International Application WO 2007/134181 published on 11/22/07 wherein a 4'-CH 2 -O-2' bicyclic nucleoside is further substituted at the 5' position with a 5'-methyl or a 5'-vinyl group.
  • carbocyclic bicyclic nucleosides along with their oligomerization and biochemical studies have also been described ( see, e.g., Srivastava et al., J. Am. Chem. Soc. 2007, 129(26), 8362-8379 ).
  • the present invention provides oligonucleotides comprising modified nucleosides.
  • modified nucleotides may include modified sugars, modified nucleobases, and/or modified linkages. The specific modifications are selected such that the resulting oligonucleotides possess desireable characteristics.
  • oligonucleotides comprise one or more RNA-like nucleosides. In certain embodiments, oligonucleotides comprise one or more DNA-like nucleotides.
  • nucleosides of the present invention comprise one or more unmodified nucleobases. In certain embodiments, nucleosides of the present invention comprise one or more modifed nucleobases.
  • modified nucleobases are selected from: universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases as defined herein.
  • nucleobases include tricyclic pyrimidines such as phenoxazine cytidine([5,4-b][1,4]benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido[5,4-b][1,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g.
  • nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in United States Patent No.
  • nucleosides may be linked together using any internucleoside linkage to form oligonucleotides.
  • the two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom.
  • Non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (-CH 2 -N(CH 3 )-O-CH 2 -), thiodiester (-O-C(O)-S-), thionocarbamate (-O-C(O)(NH)-S-); siloxane (-O-Si(H) 2 -O-); and N,N'-dimethylhydrazine (-CH 2 -N(CH 3 )-N(CH 3 )-).
  • Modified linkages compared to natural phosphodiester linkages, can be used to alter, typically increase, nuclease resistance of the oligonucleotide.
  • internucleoside linkages having a chiral atom can be prepared as a racemic mixture, or as separate enantiomers.
  • Representative chiral linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing internucleoside linkages are well known to those skilled in the art.
  • oligonucleotides described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), ⁇ or ⁇ such as for sugar anomers, or as (D) or (L) such as for amino acids etc. Included in the antisense compounds provided herein are all such possible isomers, as well as their racemic and optically pure forms.
  • Further neutral internucleoside linkages include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example: Carbohydrate Modifications in Antisense Research; Y.S. Sanghvi and P.D. Cook, Eds., ACS Symposium Series 580; Chapters 3 and 4, 40-65 ). Further neutral internucleoside linkages include nonionic linkages comprising mixed N, O, S and CH 2 component parts.
  • oligomeric compounds include nucleosides synthetically modified to induce a 3'-endo sugar conformation.
  • a nucleoside can incorporate synthetic modifications of the heterocyclic base moiety, the sugar moiety or both to induce a desired 3'-endo sugar conformation. These modified nucleosides are used to mimic RNA like nucleosides so that particular properties of an oligomeric compound can be enhanced while maintaining the desirable 3'-endo conformational geometry.
  • RNA type duplex A form helix, predominantly 3'-endo
  • duplexes composed of 2'-deoxy-2'-F-nucleosides appear efficient in triggering RNAi response in the C. elegans system.
  • Properties that are enhanced by using more stable 3'-endo nucleosides include but aren't limited to modulation of pharmacokinetic properties through modification of protein binding, protein off-rate, absorption and clearance; modulation of nuclease stability as well as chemical stability; modulation of the binding affinity and specificity of the oligomer (affinity and specificity for enzymes as well as for complementary sequences); and increasing efficacy of RNA cleavage.
  • the present invention provides oligomeric compounds having one or more nucleosides modified in such a way as to favor a C3'-endo type conformation.
  • Nucleoside conformation is influenced by various factors including substitution at the 2', 3' or 4'-positions of the pentofuranosyl sugar. Electronegative substituents generally prefer the axial positions, while sterically demanding substituents generally prefer the equatorial positions ( Principles of Nucleic Acid Structure, Wolfgang Sanger, 1984, Springer-Verlag .) Modification of the 2' position to favor the 3'-endo conformation can be achieved while maintaining the 2'-OH as a recognition element, as exemplified in Example 35, below ( Gallo et al., Tetrahedron (2001), 57, 5707-5713 . Harry-O'kuru et al., J. Org.
  • LNA locked nucleic acid
  • ENA ethylene bridged nucleic acids
  • oligomeric compounds comprise or consist of oligonucleotides.
  • such oligonucleotides comprise one or more chemical modification.
  • chemically modified oligonucleotides comprise one or more modified sugars.
  • chemically modified oligonucleotides comprise one or more modified nucleobases.
  • chemically modified oligonucleotides comprise one or more modified internucleoside linkages.
  • the chemical modifications (sugar modifications, nucleobase modifications, and/or linkage modifications) define a pattern or motif.
  • the patterns of chemical modifications of sugar moieties, internucleoside linkages, and nucleobases are each independent of one another.
  • an oligonucleotide may be described by its sugar modification motif, internucleoside linkage motif and/or nucleobase modification motif (as used herein, nucleobase modification motif describes the chemical modifications to the nucleobases independent of the sequence of nucleobases).
  • oligonucleotides comprise one or more type of modified sugar moieties and/or naturally occurring sugar moieties arranged along an oligonucleotide or region thereof in a defined pattern or sugar motif.
  • sugar motifs include but are not limited to any of the sugar modifications discussed herein.
  • the oligonucleotides comprise or consist of a region having a gapmer sugar motif, which comprises two external regions or "wings" and a central or internal region or "gap."
  • the three regions of a gapmer sugar motif (the 5'-wing, the gap, and the 3'-wing) form a contiguous sequence of nucleosides wherein at least some of the sugar moieties of the nucleosides of each of the wings differ from at least some of the sugar moieties of the nucleosides of the gap.
  • the sugar moieties of the nucleosides of each wing that are closest to the gap differ from the sugar moiety of the neighboring gap nucleosides, thus defining the boundary between the wings and the gap.
  • the sugar moieties within the gap are the same as one another.
  • the gap includes one or more nucleoside having a sugar moiety that differs from the sugar moiety of one or more other nucleosides of the gap.
  • the sugar motifs of the two wings are the same as one another (symmetric sugar gapmer).
  • the sugar motifs of the 5'-wing differs from the sugar motif of the 3'-wing (asymmetric sugar gapmer).
  • oligonucleotides comprise chemical modifications to nucleobases arranged along the oligonucleotide or region thereof in a defined pattern or nucleobases modification motif. In certain embodiments, each nucleobase is modified. In certain embodiments, none of the nucleobases is chemically modified.
  • oligonucleotides comprise a block of modified nucleobases.
  • the block is at the 3'-end of the oligonucleotide.
  • the block is within 3 nucleotides of the 3'-end of the oligonucleotide.
  • the block is at the 5'-end of the oligonucleotide. In certain embodiments the block is within 3 nucleotides of the 5'-end of the oligonucleotide.
  • nucleobase modifications are a function of the natural base at a particular position of an oligonucleotide.
  • each purine or each pyrimidine in an oligonucleotide is modified.
  • each adenine is modified.
  • each guanine is modified.
  • each thymine is modified.
  • each cytosine is modified.
  • each uracil is modified.
  • oligonucleotides comprise one or more nucleosides comprising a modified nucleobase.
  • oligonucleotides having a gapmer sugar motif comprise a nucleoside comprising a modified nucleobase.
  • one nucleoside comprising a modified nucleobases is in the central gap of an oligonucleotide having a gapmer sugar motif.
  • the sugar is an unmodified 2'deoxynucleoside.
  • the modified nucleobase is selected from: a 2-thio pyrimidine and a 5-propyne pyrimidine
  • cytosine moieties in an oligonucleotide are 5-methyl cytosine moieties.
  • 5-methyl cytosine is not a "modified nucleobase.” Accordingly, unless otherwise indicated, unmodified nucleobases include both cytosine residues having a 5-methyl and those lacking a 5 methyl. In certain embodiments, the methylation state of all or some cytosine nucleobases is specified.
  • the oligonucleotides comprise or consist of a region having a gapmer nucleoside motif, which comprises two external regions or "wings" and a central or internal region or "gap.”
  • the three regions of a gapmer nucleoside motif (the 5'-wing, the gap, and the 3'-wing) form a contiguous sequence of nucleosides wherein at least some of the sugar moieties and/or nucleobases of the nucleosides of each of the wings differ from at least some of the sugar moieties and/or nucleobase of the nucleosides of the gap.
  • the nucleosides of each wing that are closest to the gap differ from the neighboring gap nucleosides, thus defining the boundary between the wings and the gap.
  • the nucleosides within the gap are the same as one another.
  • the gap includes one or more nucleoside that differs from one or more other nucleosides of the gap.
  • the nucleoside motifs of the two wings are the same as one another (symmetric gapmer).
  • the nucleoside motifs of the 5'-wing differs from the nucleoside motif of the 3'-wing (asymmetric gapmer).
  • the 5'- wing of a gapmer consists of 1 to 6 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 5 linked nucleosides. In certain embodiments, the 5'-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 3 to 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 4 linked nucleosides.
  • the 5'- wing of a gapmer consists of 1 to 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 5'-wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 or 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 nucleoside.
  • the 5'- wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 5'-wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 6 linked nucleosides.
  • the 5'- wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least two bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least three bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least four bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least one constrained ethyl nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one LNA nucleoside.
  • each nucleoside of the 5'-wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a LNA nucleoside.
  • the 5'- wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-substituted nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-MOE nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-OMe nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a non-bicyclic modified nucleoside.
  • each nucleoside of the 5'- wing of a gapmer is a 2'-substituted nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-MOE nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-OMe nucleoside.
  • the 5'- wing of a gapmer comprises at least one 2'-deoxynucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-deoxynucleoside. In a certain embodiments, the 5'- wing of a gapmer comprises at least one ribonucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a ribonucleoside. In certain embodiments, one, more than one, or each of the nucleosides of the 5'- wing is an RNA-like nucleoside.
  • the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-OMe nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-deoxynucleoside.
  • the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-OMe nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-deoxynucleoside.
  • the 5'- wing of a gapmer has a nucleoside motif selected from among the following: ADDA; ABDAA; ABBA; ABB; ABAA; AABAA; AAABAA; AAAABAA; AAAAABAA; AAABAA; AABAA; ABAB; ABADB; ABADDB; AAABB; AAAAA; ABBDC; ABDDC; ABBDCC; ABBDDC; ABBDCC; ABBC; AA; AAA; AAAA; AAAAB; AAAAAAA; AAAAAAAA; ABBB; AB; ABAB; AAAAB; AABBB; AAAAB; and AABBB, wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type, each C is a modified nucleoside of a third type, and each D is an unmodified deoxynucleoside.
  • the 5'- wing of a gapmer has a nucleoside motif selected from among the following: AB, ABB, AAA, BBB, BBBAA, AAB, BAA, BBAA, AABB, AAAB, ABBW, ABBWW, ABBB, ABBBB, ABAB, ABABAB, ABABBB, ABABAA, AAABB, AAAABB, AABB, AAAAB, AABBB, ABBBB, BBBBB, AAABW, AAAAA, BBBBAA, and AAABW; wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type, and each W is a modified nucleoside of either the first type, the second type or a third type.
  • the 5'- wing of a gapmer has a nucleoside motif selected from among the following: ABB; ABAA; AABAA; AAABAA; ABAB; ABADB; AAABB; AAAAA; AA; AAA; AAAA; AAAAB; ABBB; AB; and ABAB; wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type, and each W is a modified nucleoside of either the first type, the second type or a third type.
  • an oligonucleotide comprises any 5'-wing motif provided herein.
  • the oligonucleotide is a 5'-hemimer (does not comprise a 3'-wing).
  • such an oligonucleotide is a gapmer.
  • the 3'-wing of the gapmer may comprise any nucleoside motif.
  • the 5'- wing of a gapmer has a sugar motif selected from among those listed in the following non-limiting tables: Table 1 Certain 5'-Wing Sugar Motifs Certain 5'-Wing Sugar Motifs AAAAA ABCBB BABCC BCBBA CBACC AAAAB ABCBC BACAA BCBBB CBBAA AAAAC ABCCA BACAB BCBBC CBBAB AAABA ABCCB BACAC BCBCA CBBAC AAABB ABCCC BACBA BCBCB CBBBA AAABC ACAAA BACBB BCBCC CBBBB AAACA ACAAB BACBC BCCAA CBBBC AAACB ACAAC BACCA BCCAB CBBCA AAACC ACABA BACCB BCCAC CBBCB AABAA ACABB BACCC BCCBA CBBCC AABAB ACABC BBAAA BCCBB CBCAA AABAC ACACA BBAAB BCCBC CBCAB AABBA ACACB BBAAC BCCCA CBCAC AABBB ACACC BB BCCCB CBCAA AABAC ACACA BBAAB
  • each A, each B, and each C located at the 3'-most 5'-wing nucleoside is a modified nucleoside.
  • the 5'-wing motif is selected from among ABB, BBB, and CBB, wherein the underlined nucleoside represents the 3'-most 5'-wing nucleoside and wherein the underlined nucleoside is a modified nucleoside.
  • the the 3'-most 5'-wing nucleoside comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • the the 3'-most 5'-wing nucleoside comprises a bicyclic sugar moiety selected from among cEt and LNA. In certain embodiments, the the 3'-most 5'-wing nucleoside comprises cEt. In certain embodiments, the the 3'-most 5'-wing nucleoside comprises LNA.
  • each A comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each A comprises a modified sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety selected from among F, ara-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each A comprises a bicyclic sugar moiety. In certain embodiments, each A comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each A comprises a modified nucleobase. In certain embodiments, each A comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside. In certain embodiments, each A comprises an HNA. In certain embodiments, each A comprises a F-HNA. In certain embodiments, each A comprises a 5'-substituted sugar moiety selected from among 5'-Me DNA, and 5'- (R) -Me DNA.
  • each B comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each B comprises a modified sugar moiety. In certain embodiments, each B comprises a 2'-substituted sugar moiety. In certain embodiments, each B comprises a 2'-subsituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each B comprises a bicyclic sugar moiety. In certain embodiments, each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each B comprises a modified nucleobase. In certain embodiments, each B comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne urindine nucleoside. In certain embodiments, each B comprises an HNA. In certain embodiments, each B comprises a F-HNA. In certain embodiments, each B comprises a 5'-substituted sugar moiety selected from among 5'-Me DNA, and 5'- (R) -Me DNA.
  • each A comprises a 2'-substituted sugar moiety selected from among F, ara-F, OCH 3 and O(CH 2 ) 2 -OCH 3 and each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each A comprises O(CH 2 ) 2 -OCH 3 and each B comprises cEt.
  • each C comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each C comprises a modified sugar moiety. In certain embodiments, each C comprises a 2'-substituted sugar moiety. In certain embodiments, each C comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each C comprises a 5'-substituted sugar moiety. In certain embodiments, each C comprises a 5'-substituted sugar moiety selected from among 5'-Me DNA, and 5'- (R) -Me DNA.
  • each C comprises a bicyclic sugar moiety. In certain embodiments, each C comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA. In certain embodiments, each C comprises a modified nucleobase. In certain embodiments, each C comprises a modified nucleobase selected from among 2-thio-thymidine and 5-propyne uridine. In certain embodiments, each C comprises a 2-thio-thymidine nucleoside. In certain embodiments, each C comprises an HNA. In certain embodiments, each C comprises an F-HNA.
  • the 3'- wing of a gapmer consists of 1 to 6 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 5 linked nucleosides. In certain embodiments, the 3'-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 3 to 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 4 linked nucleosides.
  • the 3'- wing of a gapmer consists of 1 to 3 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 3'-wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 or 3 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 nucleoside.
  • the 3'- wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 3'-wing of a gapmer consists of 3linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 6 linked nucleosides.
  • the 3'- wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one constrained ethyl nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one LNA nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a LNA nucleoside.
  • the 3'- wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least two non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least three non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least four non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-substituted nucleoside.
  • the 3'- wing of a gapmer comprises at least one 2'-MOE nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-OMe nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a non-bicyclic modified nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-substituted nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-MOE nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-OMe nucleoside.
  • the 3'- wing of a gapmer comprises at least one 2'-deoxynucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-deoxynucleoside. In a certain embodiments, the 3'- wing of a gapmer comprises at least one ribonucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a ribonucleoside. In certain embodiments, one, more than one, or each of the nucleosides of the 5'- wing is an RNA-like nucleoside.
  • the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-OMe nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-deoxynucleoside.
  • the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-OMe nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-deoxynucleoside.
  • the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-OMe nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-deoxynucleoside.
  • the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside.
  • the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside.
  • the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside.
  • the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-OMe nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-OMe nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside, at least one 2'-OMe nucleoside, and at least one 2'-deoxynucleoside.
  • the 3'- wing of a gapmer has a nucleoside motif selected from among the following: ABB, ABAA, AAABAA, AAAAABAA, AABAA, AAAABAA, AAABAA, ABAB, AAAAA, AAABB, AAAAAAAA, AAAAAAA, AAAAAA, AAAAB, AAAA, AAA, AA, AB, ABBB, ABAB, AABBB; wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type.
  • an oligonucleotide comprises any 3'-wing motif provided herein.
  • the oligonucleotide is a 3'-hemimer (does not comprise a 5'-wing). In certain embodiments, such an oligonucleotide is a gapmer. In certain such embodiments, the 5'-wing of the gapmer may comprise any nucleoside motif.
  • the 3'- wing of a gapmer has a nucleoside motif selected from among the following: BBA, AAB, AAA, BBB, BBAA, AABB, WBBA, WAAB, BBBA, BBBBA, BBBB, BBBBBA, ABBBBB, BBAAA, AABBB, BBBAA, BBBBA, BBBBB, BABA, AAAAA, BBAAAA, AABBBB, BAAAA, and ABBBB, wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type, and each W is a modified nucleoside of either the first type, the second type or a third type.
  • the 3'- wing of a gapmer has a sugar motif selected from among those listed in the following non-limiting tables: Table 3 Certain 3'-Wing Sugar Motifs Certain 3'-Wing Sugar Motifs AAAAA ABCBB BABCC BCBBA CBACC AAAAB ABCBC BACAA BCBBB CBBAA AAAAC ABCCA BACAB BCBBC CBBAB AAABA ABCCB BACAC BCBCA CBBAC AAABB ABCCC BACBA BCBCB CBBBA AAABC ACAAA BACBB BCBCC CBBBB AAACA ACAAB BACBC BCCAA CBBBC AAACB ACAAC BACCA BCCAB CBBCA AAACC ACABA BACCB BCCAC CBBCB AABAA ACABB BACCC BCCBA CBBCC AABAB ACABC BBAAA BCCBB CBCAA AABAC ACACA BBAAB BCCBC CBCAB AABBA ACACB BBAAC BCCCA CBCAC AABBB ACACC BB BCCCB CBCAA AABAC ACACA BBAAB
  • each A, each B, and each C located at the 5'-most 3'-wing region nucleoside is a modified nucleoside.
  • the 3'-wing motif is selected from among ABB, BBB, and CBB, wherein the underlined nucleoside represents the the 5'-most 3'-wing region nucleoside and wherein the underlined nucleoside is a modified nucleoside.
  • each A comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each A comprises a modified sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety selected from among F, ara-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each A comprises a bicyclic sugar moiety. In certain embodiments, each A comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each A comprises a modified nucleobase. In certain embodiments, each A comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside. In certain embodiments, each A comprises a 5'-substituted sugar moiety selected from among 5'-Me DNA, and 5'- (R) -Me DNA.
  • each B comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each B comprises a modified sugar moiety. In certain embodiments, each B comprises a 2'-substituted sugar moiety. In certain embodiments, each B comprises a 2'-subsituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each B comprises a bicyclic sugar moiety. In certain embodiments, each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each B comprises a modified nucleobase. In certain embodiments, each B comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne urindine nucleoside. In certain embodiments, each B comprises an HNA. In certain embodiments, each B comprises an F-HNA. In certain embodiments, each B comprises a 5'-substituted sugar moiety selected from among 5'-Me DNA, and 5'- (R) -Me DNA.
  • each A comprises a 2'-substituted sugar moiety selected from among F, ara-F, OCH 3 and O(CH 2 ) 2 -OCH 3 and each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each A comprises O(CH 2 ) 2 -OCH 3 and each B comprises cEt.
  • each C comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each C comprises a modified sugar moiety. In certain embodiments, each C comprises a 2'-substituted sugar moiety. In certain embodiments, each C comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each C comprises a 5'-substituted sugar moiety. In certain embodiments, each C comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me. In certain embodiments, each C comprises a bicyclic sugar moiety.
  • each C comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each C comprises a modified nucleobase.
  • each C comprises a modified nucleobase selected from among 2-thio-thymidine and 5-propyne uridine.
  • each C comprises a 2-thio-thymidine nucleoside.
  • each C comprises an HNA.
  • each C comprises an F-HNA.
  • the gap of a gapmer consists of 6 to 20 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 15 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 12 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 or 7 linked nucleosides.
  • the gap of a gapmer consists of 7 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 or 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 or 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 linked nucleosides.
  • the gap of a gapmer consists of 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 11 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 12 linked nucleosides.
  • each nucleoside of the gap of a gapmer is a 2'-deoxynucleoside.
  • the gap comprises one or more modified nucleosides.
  • each nucleoside of the gap of a gapmer is a 2'-deoxynucleoside or is a modified nucleoside that is "DNA-like.”
  • DNA-like means that the nucleoside has similar characteristics to DNA, such that a duplex comprising the gapmer and an RNA molecule is capable of activating RNase H. For example, under certain conditions, 2'-(ara)-F have been shown to support RNase H activation, and thus is DNA-like.
  • one or more nucleosides of the gap of a gapmer is not a 2'-deoxynucleoside and is not DNA-like. In certain such embodiments, the gapmer nonetheless supports RNase H activation (e.g., by virtue of the number or placement of the non-DNA nucleosides).
  • gaps comprise a stretch of unmodified 2'-deoxynucleoside interrupted by one or more modified nucleosides, thus resulting in three sub-regions (two stretches of one or more 2'-deoxynucleosides and a stretch of one or more interrupting modified nucleosides).
  • no stretch of unmodified 2'-deoxynucleosides is longer than 5, 6, or 7 nucleosides.
  • such short stretches is achieved by using short gap regions.
  • short stretches are achieved by interrupting a longer gap region.
  • the gap comprises one or more modified nucleosides. In certain embodiments, the gap comprises one or more modified nucleosides selected from among cEt, FHNA, LNA, and 2-thio-thymidine. In certain embodiments, the gap comprises one modified nucleoside. In certain embodiments, the gap comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me. In certain embodiments, the gap comprises two modified nucleosides. In certain embodiments, the gap comprises three modified nucleosides. In certain embodiments, the gap comprises four modified nucleosides. In certain embodiments, the gap comprises two or more modified nucleosides and each modified nucleoside is the same. In certain embodiments, the gap comprises two or more modified nucleosides and each modified nucleoside is different.
  • the gap comprises one or more modified linkages. In certain embodiments, the gap comprises one or more methyl phosphonate linkages. In certain embodiments the gap comprises two or more modified linkages. In certain embodiments, the gap comprises one or more modified linkages and one or more modified nucleosides. In certain embodiments, the gap comprises one modified linkage and one modified nucleoside. In certain embodiments, the gap comprises two modified linkages and two or more modified nucleosides.
  • the gap comprises a nucleoside motif selected from among the following: DDDDXDDDDD; DDDDDXDDDDD; DDDXDDDDD; DDDDXDDDDDD; DDXDDDDDD; DDDXDDDDDD; DDXDDDDD; DDXDDDDD; DDXDDDXDDD; DDDXDDDXDDD; DDXDDDXDD; DDXDDDDXDD; DDXDDDDXDD; DDXDDDDXDD; DDXDDDDXDD; DDXDDDDXDD; DXDDDDXDDD; DDDDXDDD; DXDDDDXDDD; DDDDXDDD; DDDDDXDDD; DDDDDXDDD; DDDDDXDDDDD; DDDDXDDDDD; DDDDXDDD; DDDXDDDDDDD; DDDDXDDDDD; DDDDXDDD; DDDXDDDDDDD; DDDDXDDD; DDDDXDDD;
  • the gap comprises a nucleoside motif selected from among the following: DDDDDDDDD; DXDDDDD; DDXDDDD; DDDXDDDDD; DDDDXDDDD; DDDDDXDDD; DDDDDDXDDDD; DDDDDDDXD; DDDDDDXDDDD; DDXXXDDDD; DDDDXXDDDDD; DDDDXXDDDDD; DDDDXXDDD; DDDDDXXDD; DDDDDXDD; DXDDDDXDD; DXDDXDDDD; DXDDXDDDD; DDXDDDDXD; DDXDDDDXDD; DDXDDXDD; DDXDDXDD; DDXDDDXDD; DDXDXDDDD; DDXDXDDDD; DDXDXDDDD; DDXDXDDDD; DDXDXDDDD; DDXDXDDDD; DDXDXDDDD; DDXDXDDDD; DDXDXDD
  • the gap comprises a nucleoside motif selected from among the following: DDDDXDDDD, DXDDDDD, DXXDDDDDD, DDXDDDDDD, DDDXDDDDD, DDDDXDDDD, DDDDDXDDD, DDDDDDXDD, and DDDDDDDXD, wherein each D is an unmodified deoxynucleoside; and each X is a modified nucleoside or a substituted sugar moiety.
  • the gap comprises a nucleoside motif selected from among the following: DDDDDDDD, DXDDDD, DDXDDDDD, DDDXDDDD, DDDDXDDD, DDDDDXDD, DDDDDDXD, DXDDDDXD, DXDDDDXDD, DXDDXDDDD, DDXXDDDD, DDXDDXDD, DDXDDXDD, DDXDDXDD, DDXDDXDD, DDXDDXDD, DXDDXD, DDDXXD, DDDXDXDD, DDDXDDXD, DDDDXXDD, DDDDXDXD, and DDDDDXXD, wherein each D is an unmodified deoxynucleoside; and each X is a modified nucleoside or a substituted sugar moiety.
  • the gap comprises a nucleoside motif selected from among the following: DXDDDDD, DDXDDDD, DDDXDDD, DDDDXDD, DDDDDXD, DXDDDXD, DXDDXDD, DXDXDDDD, DDXXDDD, DDXDDXD, DDXDDXD, DDDXXDD, DDDXDXD, and DDDDXXD, wherein each D is an unmodified deoxynucleoside; and each X is a modified nucleoside or a substituted sugar moiety.
  • the gap comprises a nucleoside motif selected from among the following: DXDDDD, DDXDDD, DDDXDD, DDDDXD, DXXDDD, DXDXDD, DXDDXD, DDXXDD, DDXDXD, and DDDXXD, wherein each D is an unmodified deoxynucleoside; and each X is a modified nucleoside or a substituted sugar moiety.
  • the gap comprises a nucleoside motif selected from among the following: DXDDDD, DDXDDD, DDDXDD, DDDDXD, DXDDDDD, DDXDDDD, DDDDDXD, DXDDDDDD, DDXDDDDD, DDDXDDDD, DDDDXDDD, DDDDDXDD, DDDDDDXD, DXDDDDDDD; DDXDDDDDD, DDDXDDDDD, DDDDXDD, DDDDDXDD, DDDDXDD, DDDDDDDXD, DXDDDDDDDD, DDXDDDDD, DDDXDDDDDDDD, DDXDDDDDDD, DDDXDDDDDDDD, DDDDXDDDDDDD, DDDXDDDDDDDD, DDDDXDDDDDDD, DDDDDXDDDDDD, DDDDXDDDDDDD, DDDDDXDDDDDD, DDDDXDDDDDDD, DDDDDXDDDDDD, DDDDXDDDDD
  • each X comprises an unmodified 2'-deoxyfuranose sugar moiety. In certain embodiments, each X comprises a modified sugar moiety. In certain embodiments, each X comprises a 2'-substituted sugar moiety. In certain embodiments, each X comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each X comprises a 5'-substituted sugar moiety. In certain embodiments, each X comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me.
  • each X comprises a bicyclic sugar moiety. In certain embodiments, each X comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA. In certain embodiments, each X comprises a modified nucleobase. In certain embodiments, each X comprises a modified nucleobase selected from among 2-thio-thymidine and 5-propyne uridine. In certain embodiments, each X comprises a 2-thio-thymidine nucleoside. In certain embodiments, each X comprises an HNA. In certain embodiments, each C comprises an F-HNA. In certain embodiments, X represents the location of a single differentiating nucleobase.
  • a gapmer comprises a 5'-wing, a gap, and a 3' wing, wherein the 5'-wing, gap, and 3' wing are independently selected from among those discussed above.
  • a gapmer has a 5'-wing, a gap, and a 3'-wing having features selected from among any of those listed in the tables above and any 5'-wing may be paired with any gap and any 3'-wing.
  • a 5'-wing may comprise AAABB
  • a 3'-wing may comprise BBA
  • the gap may comprise DDDDDDD.
  • a gapmer comprises a 5'-wing, a gap, and a 3' wing, wherein the 5'-wing, gap, and 3' wing are independently selected from among those discussed above.
  • a gapmer has a 5'-wing, a gap, and a 3'-wing having features selected from among those listed in the following non-limiting tables: Table 6 Certain Gapmer Nucleoside Motifs 5'-wing region Central gap region 3'-wing region ADDA DDDD ABB ABBA DDDADDDD ABAA AAAAAAA DDDDDDDDD AAA AAAAABB DDDDDDDD BBAAAAA ABB DDDDADDDD ABB ABB DDDDBDDDD BBA ABB DDDDDDDDD BBA AABAA DDDDDDD AABAA ABB DDDDDD AABAA AAABAA DDDDDDDDD AAABAA AAABAA DDDDDDDDD AAB ABAB DDDDDDDDD ABAB AAABB DDDDDDD BBA ABA
  • each A comprises a modified sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety selected from among F, ara-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each A comprises a bicyclic sugar moiety. In certain embodiments, each A comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA. In certain embodiments, each A comprises a modified nucleobase.
  • each A comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside.
  • each A comprises an HNA.
  • each A comprises an F-HNA.
  • each A comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me.
  • each B comprises a modified sugar moiety. In certain embodiments, each B comprises a 2'-substituted sugar moiety. In certain embodiments, each B comprises a 2'-subsituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each B comprises a bicyclic sugar moiety. In certain embodiments, each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA. In certain embodiments, each B comprises a modified nucleobase.
  • each B comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne urindine nucleoside.
  • each B comprises an HNA.
  • each B comprises an F-HNA.
  • each B comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me.
  • each C comprises a modified sugar moiety. In certain embodiments, each C comprises a 2'-substituted sugar moiety. In certain embodiments, each C comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each C comprises a 5'-substituted sugar moiety. In certain embodiments, each C comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me. In certain embodiments, each C comprises a bicyclic sugar moiety.
  • each C comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each C comprises a modified nucleobase.
  • each C comprises a modified nucleobase selected from among 2-thio-thymidine and 5-propyne uridine.
  • each C comprises a 2-thio-thymidine nucleoside.
  • each C comprises an HNA.
  • each C comprises an F-HNA.
  • each W comprises a modified sugar moiety. In certain embodiments, each W comprises a 2'-substituted sugar moiety. In certain embodiments, each W comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each W comprises a 5'-substituted sugar moiety. In certain embodiments, each W comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me. In certain embodiments, each W comprises a bicyclic sugar moiety.
  • each W comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each W comprises a sugar surrogate.
  • each W comprises a sugar surrogate selected from among HNA and F-HNA.
  • each W comprises a 2-thio-thymidine nucleoside.
  • At least one of A or B comprises a bicyclic sugar moiety, and the other comprises a 2'-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-substituted sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-substituted sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-MOE sugar moiety. In certain embodiments, one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-MOE sugar moiety. In certain embodiments, one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-MOE sugar moiety. In certain embodiments, one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-F sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-F sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-F sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-(ara)-F sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-(ara)-F sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-(ara)-F sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-substituted sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-substituted sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-substituted sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-substituted sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-MOE sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-MOE sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-MOE sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-MOE sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-F sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-F sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-F sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-F sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-(ara)-F sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-(ara)-F sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-(ara)-F sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-(ara)-F sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2'-MOE sugar moiety.
  • B is an LNA nucleoside and A comprises a 2'-MOE sugar moiety.
  • B is a cEt nucleoside and A comprises a 2'-MOE sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2'-MOE sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2'-F sugar moiety.
  • B is an LNA nucleoside and A comprises a 2'-F sugar moiety.
  • B is a cEt nucleoside and A comprises a 2'-F sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2'-F sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2'-(ara)-F sugar moiety.
  • B is an LNA nucleoside and A comprises a 2'-(ara)-F sugar moiety.
  • B is a cEt nucleoside and A comprises a 2'-(ara)-F sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2'-(ara)-F sugar moiety.
  • At least one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-substituted sugar moiety and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and C comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises 2-thio-thymidine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and C comprises a 5-propyne uridine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and C comprises a 5-propyne uridine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and C comprises a 5-propyne uridine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar HNA surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside
  • another of A or B comprises a 2'-(ara)-F sugar moiety
  • W comprises a 5'- (R) -Me DNA sugar moiety.
  • At least two of A, B or W comprises a 2'-substituted sugar moiety, and the other comprises a bicyclic sugar moiety. In certain embodiments, at least two of A, B or W comprises a bicyclic sugar moiety, and the other comprises a 2'-substituted sugar moiety.
  • a gapmer has a sugar motif other than: E-K-K-(D) 9 -K-K-E; E-E-E-E-K-(D) 9 -E-E-E-E; E-K-K-K-(D) 9 -K-K-E; K-E-E-K-(D) 9 -K-E-E-K; K-D-D-K-(D) 9 -K-D-D-K; K-E-K-E-K-(D) 9 -K-E-K-E-K; K-D-K-D-K-(D) 9 -K-D-K-D-K; E-K-E-K-(D) 9 -K-E-K-E; E-E-E-E-E-K-(D) 8 -E-E-E-E-E; or E-K-E-K-E-(D) 9 -E-K-E-K-E-E-(D) 9
  • a gapmer comprises a A-(D) 4 -A-(D) 4 -A-(D) 4 -AA motif. In certain embodiments a gapmer comprises a B-(D) 4 -A-(D) 4 -A-(D) 4 -AA motif. In certain embodiments a gapmer comprises a A-(D) 4 -B-(D) 4 -A-(D) 4 -AA motif. In certain embodiments a gapmer comprises a A-(D) 4 -A-(D) 4 -B-(D) 4 -AA motif. In certain embodiments a gapmer comprises a A-(D) 4 -A-(D) 4 -A-(D) 4 -BA motif.
  • a gapmer comprises a A-(D) 4 -A-(D) 4 -A-(D) 4 -BB motif. In certain embodiments a gapmer comprises a K-(D) 4 -K-(D) 4 -K-(D) 4 -K-E motif.
  • oligonucleotides comprise modified internucleoside linkages arranged along the oligonucleotide or region thereof in a defined pattern or modified internucleoside linkage motif.
  • internucleoside linkages are arranged in a gapped motif, as described above for nucleoside motif.
  • the internucleoside linkages in each of two wing regions are different from the internucleoside linkages in the gap region.
  • the internucleoside linkages in the wings are phosphodiester and the internucleoside linkages in the gap are phosphorothioate.
  • the nucleoside motif is independently selected, so such oligonucleotides having a gapped internucleoside linkage motif may or may not have a gapped nucleoside motif and if it does have a gapped nucleoside motif, the wing and gap lengths may or may not be the same.
  • oligonucleotides comprise a region having an alternating internucleoside linkage motif. In certain embodiments, oligonucleotides of the present invention comprise a region of uniformly modified internucleoside linkages. In certain such embodiments, the oligonucleotide comprises a region that is uniformly linked by phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide is uniformly linked by phosphorothioate. In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate. In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate and at least one internucleoside linkage is phosphorothioate.
  • the oligonucleotide comprises at least 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages.
  • the oligonucleotide comprises at least one block of at least 10 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least block of at least one 12 consecutive phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide.
  • oligonucleotides comprise one or more methylphosponate linkages.
  • oligonucleotides having a gapmer nucleoside motif comprise a linkage motif comprising all phosphorothioate linkages except for one or two methylphosponate linkages.
  • one methylphosponate linkage is in the central gap of an oligonucleotide having a gapmer nucleoside motif.
  • Modification motifs define oligonucleotides by nucleoside motif (sugar motif and nucleobase motif) and linkage motif.
  • certain oligonucleotides have the following modification motif: A s A s A s D s D s D s D s ( N D) s D s D s D s D s B s B; wherein each A is a modified nucleoside comprising a 2'-substituted sugar moiety; each D is an unmodified 2'-deoxynucleoside; each B is a modified nucleoside comprising a bicyclic sugar moiety; N D is a modified nucleoside comprising a modified nucleobase; and s is a phosphorothioate internucleoside linkage.
  • the sugar motif is a gapmer motif.
  • the nucleobase modification motif is a single modified nucleobase at 8 th nucleoside from the 5'-end.
  • the nucleoside motif is an interrupted gapmer where the gap of the sugar modified gapmer is interrupted by a nucleoside comprising a modified nucleobase.
  • the linkage motif is uniform phosphorothioate.
  • each A comprises a modified sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety. In certain embodiments, each A comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each A comprises a bicyclic sugar moiety. In certain embodiments, each A comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA. In certain embodiments, each A comprises a modified nucleobase.
  • each A comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside.
  • each B comprises a modified sugar moiety.
  • each B comprises a 2'-substituted sugar moiety.
  • each B comprises a 2'-subsituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 .
  • each B comprises a bicyclic sugar moiety.
  • each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each B comprises a modified nucleobase.
  • each B comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne urindine nucleoside.
  • each A comprises an HNA.
  • each A comprises an F-HNA.
  • each W comprises a modified sugar moiety. In certain embodiments, each W comprises a 2'-substituted sugar moiety. In certain embodiments, each W comprises a 2'-substituted sugar moiety selected from among F, (ara)-F, OCH 3 and O(CH 2 ) 2 -OCH 3 . In certain embodiments, each W comprises a 5'-substituted sugar moiety. In certain embodiments, each W comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'- (R) -Me. In certain embodiments, each W comprises a bicyclic sugar moiety.
  • each W comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2'-thio LNA.
  • each W comprises a sugar surrogate.
  • each W comprises a sugar surrogate selected from among HNA and F-HNA.
  • At least one of A or B comprises a bicyclic sugar moiety, and the other comprises a 2'-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-substituted sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-substituted sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-MOE sugar moiety. In certain embodiments, one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-MOE sugar moiety. In certain embodiments, one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-MOE sugar moiety. In certain embodiments, one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-F sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-F sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-F sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2'-(ara)-F sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2'-(ara)-F sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2'-(ara)-F sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-substituted sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-substituted sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-substituted sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-substituted sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-MOE sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-MOE sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-MOE sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-MOE sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-F sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-F sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-F sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-F sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2'-(ara)-F sugar moiety.
  • A is an LNA nucleoside and B comprises a 2'-(ara)-F sugar moiety.
  • A is a cEt nucleoside and B comprises a 2'-(ara)-F sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2'-(ara)-F sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2'-MOE sugar moiety.
  • B is an LNA nucleoside and A comprises a 2'-MOE sugar moiety.
  • B is a cEt nucleoside and A comprises a 2'-MOE sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2'-MOE sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2'-F sugar moiety.
  • B is an LNA nucleoside and A comprises a 2'-F sugar moiety.
  • B is a cEt nucleoside and A comprises a 2'-F sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2'-F sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2'-(ara)-F sugar moiety.
  • B is an LNA nucleoside and A comprises a 2'-(ara)-F sugar moiety.
  • B is a cEt nucleoside and A comprises a 2'-(ara)-F sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2'-(ara)-F sugar moiety.
  • At least one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-substituted sugar moiety and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and C comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a modified nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-substituted sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 2-thio-thymidine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises 2-thio-thymidine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and C comprises a 5-propyne uridine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and C comprises a 5-propyne uridine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and C comprises a 5-propyne uridine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5-propyne uridine nucleobase.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a sugar HNA surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a F-HNA sugar surrogate.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'-Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-MOE sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside, another of A or B comprises a 2'-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B comprises a bicyclic sugar moiety, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an LNA nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is a cEt nucleoside, another of A or B comprises a 2'-(ara)-F sugar moiety, and W comprises a 5'- (R) -Me DNA sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside
  • another of A or B comprises a 2'-(ara)-F sugar moiety
  • W comprises a 5'- (R) -Me DNA sugar moiety.
  • At least two of A, B or W comprises a 2'-substituted sugar moiety, and the other comprises a bicyclic sugar moiety. In certain embodiments, at least two of A, B or W comprises a bicyclic sugar moiety, and the other comprises a 2'-substituted sugar moiety.
  • the present invention provides oligomeric compounds including oligonucleotides of any of a variety of ranges of lengths.
  • the invention provides oligomeric compounds or oligonucleotides consisting of X to Y linked nucleosides, where X represents the fewest number of nucleosides in the range and Y represents the largest number of nucleosides in the range.
  • X and Y are each independently selected from 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, and 50; provided that X ⁇ Y.
  • the invention provides oligomeric compounds which comprise oligonucleotides consisting of 8 to 9, 8 to 10, 8 to 11, 8 to 12, 8 to 13, 8 to 14, 8 to 15, 8 to 16, 8 to 17, 8 to 18, 8 to 19, 8 to 20, 8 to 21, 8 to 22, 8 to 23, 8 to 24, 8 to 25, 8 to 26, 8 to 27, 8 to 28, 8 to 29, 8 to 30, 9 to 10, 9 to 11, 9 to 12, 9 to 13, 9 to 14, 9 to 15, 9 to 16, 9 to 17, 9 to 18, 9 to 19, 9 to 20, 9 to 21, 9 to 22, 9 to 23, 9 to 24, 9 to 25, 9 to 26, 9 to 27, 9 to 28, 9 to 29, 9 to 30, 10 to 11, 10 to 12, 10 to 13, 10 to 14, 10 to 15, 10 to 16, 10 to 17, 10 to 18, 10 to 19, 10 to 20, 10 to 21, 10 to 22, 10 to 23, 10 to 24, 10 to 25, 10 to 26, 10 to 27, 10 to 28, 10 to 29, 10 to 30, 11 to 12, 11 to 13, 11 to 14, 11 to 15, 11 to 16, 11 to 17, 11 to 18, 11 to 19, 11 to 20, 11 to 21, 11 to 22, 11 to 23, 11 to 15, 11 to 16, 11 to
  • the oligomeric compound or oligonucleotide may, nonetheless further comprise additional other substituents.
  • an oligonucleotide comprising 8-30 nucleosides excludes oligonucleotides having 31 nucleosides, but, unless otherwise indicated, such an oligonucleotide may further comprise, for example one or more conjugates, terminal groups, or other substituents.
  • a gapmer oligonucleotide has any of the above lengths.
  • an oligonucleotide is described by an overall length range and by regions having specified lengths, and where the sum of specified lengths of the regions is less than the upper limit of the overall length range, the oligonucleotide may have additional nucleosides, beyond those of the specified regions, provided that the total number of nucleosides does not exceed the upper limit of the overall length range.
  • oligonucleotides of the present invention are characterized by their modification motif and overall length. In certain embodiments, such parameters are each independent of one another.
  • each internucleoside linkage of an oligonucleotide having a gapmer sugar motif may be modified or unmodified and may or may not follow the gapmer modification pattern of the sugar modifications.
  • the internucleoside linkages within the wing regions of a sugar-gapmer may be the same or different from one another and may be the same or different from the internucleoside linkages of the gap region.
  • sugar-gapmer oligonucleotides may comprise one or more modified nucleobase independent of the gapmer pattern of the sugar modifications.
  • oligonucleotide motifs may be combined to create a variety of oligonucleotides.
  • oligonucleotide or oligomeric compound is silent with respect to one or more parameter, such parameter is not limited.
  • an oligomeric compound described only as having a gapmer sugar motif without further description may have any length, internucleoside linkage motif, and nucleobase modification motif. Unless otherwise indicated, all chemical modifications are independent of nucleobase sequence.
  • oligomeric compounds are modified by attachment of one or more conjugate groups.
  • conjugate groups modify one or more properties of the attached oligomeric compound including but not limited to pharmacodynamics, pharmacokinetics, stability, binding, absorption, cellular distribution, cellular uptake, charge and clearance.
  • Conjugate groups are routinely used in the chemical arts and are linked directly or via an optional conjugate linking moiety or conjugate linking group to a parent compound such as an oligomeric compound, such as an oligonucleotide.
  • Conjugate groups includes without limitation, intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, thioethers, polyethers, cholesterols, thiocholesterols, cholic acid moieties, folate, lipids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins and dyes.
  • Certain conjugate groups have been described previously, for example: cholesterol moiety ( Letsinger et al., Proc. Natl. Acad. Sci.
  • cholic acid Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060
  • a thioether e.g., hexyl-S-tritylthiol
  • Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770 Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770
  • a thiocholesterol Oberhauser et al., Nucl.
  • Acids Res., 1990, 18, 3777-3783 a polyamine or a polyethylene glycol chain ( Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973 ), or adamantane acetic acid ( Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654 ), a palmityl moiety ( Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237 ), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety ( Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937 ).
  • a conjugate group comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, ( S )-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • active drug substance for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, ( S )-(+)-pranoprofen, carprofen, dansy
  • conjugate groups are directly attached to oligonucleotides in oligomeric compounds.
  • conjugate groups are attached to oligonucleotides by a conjugate linking group.
  • conjugate linking groups including, but not limited to, bifunctional linking moieties such as those known in the art are amenable to the compounds provided herein.
  • Conjugate linking groups are useful for attachment of conjugate groups, such as chemical stabilizing groups, functional groups, reporter groups and other groups to selective sites in a parent compound such as for example an oligomeric compound.
  • a bifunctional linking moiety comprises a hydrocarbyl moiety having two functional groups.
  • One of the functional groups is selected to bind to a parent molecule or compound of interest and the other is selected to bind essentially any selected group such as chemical functional group or a conjugate group.
  • the conjugate linker comprises a chain structure or an oligomer of repeating units such as ethylene glycol or amino acid units.
  • functional groups that are routinely used in a bifunctional linking moiety include, but are not limited to, electrophiles for reacting with nucleophilic groups and nucleophiles for reacting with electrophilic groups.
  • bifunctional linking moieties include amino, hydroxyl, carboxylic acid, thiol, unsaturations (e.g., double or triple bonds), and the like.
  • conjugate linking moieties include pyrrolidine, 8-amino-3,6-dioxaoctanoic acid (ADO), succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC) and 6-aminohexanoic acid (AHEX or AHA).
  • ADO 8-amino-3,6-dioxaoctanoic acid
  • SMCC succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate
  • AHEX or AHA 6-aminohexanoic acid
  • linking groups include, but are not limited to, substituted C 1 -C 10 alkyl, substituted or unsubstituted C 2 -C 10 alkenyl or substituted or unsubstituted C 2 -C 10 alkynyl, wherein a nonlimiting list of preferred substituent groups includes hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro, thiol, thioalkoxy, halogen, alkyl, aryl, alkenyl and alkynyl.
  • Conjugate groups may be attached to either or both ends of an oligonucleotide (terminal conjugate groups) and/or at any internal position.
  • conjugate groups are at the 3'-end of an oligonucleotide of an oligomeric compound. In certain embodiments, conjugate groups are near the 3'-end. In certain embodiments, conjugates are attached at the 3'end of an oligomeric compound, but before one or more terminal group nucleosides. In certain embodiments, conjugate groups are placed within a terminal group. In certain embodiments, the present invention provides oligomeric compounds. In certain embodiments, oligomeric compounds comprise an oligonucleotide. In certain embodiments, an oligomeric compound comprises an oligonucleotide and one or more conjugate and/or terminal groups.
  • conjugate and/or terminal groups may be added to oligonucleotides having any of the motifs discussed above.
  • an oligomeric compound comprising an oligonucleotide having region of alternating nucleosides may comprise a terminal group.
  • oligomeric compounds provided herein are antisense compounds. Such antisense compounds are capable of hybridizing to a target nucleic acid, resulting in at least one antisense activity. In certain embodiments, antisense compounds specifically hybridize to one or more target nucleic acid.
  • a specifically hybridizing antisense compound has a nucleobase sequence comprising a region having sufficient complementarity to a target nucleic acid to allow hybridization and result in antisense activity and insufficient complementarity to any non-target so as to avoid non-specific hybridization to any non-target nucleic acid sequences under conditions in which specific hybridization is desired (e.g., under physiological conditions for in vivo or therapeutic uses, and under conditions in which assays are performed in the case of in vitro assays).
  • the present invention provides antisense compounds comprising oligonucleotides that are fully complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain embodiments, oligonucleotides are 99% complementary to the target nucleic acid. In certain embodiments, oligonucleotides are 95% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 90% complementary to the target nucleic acid.
  • such oligonucleotides are 85% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 80% complementary to the target nucleic acid. In certain embodiments, an antisense compound comprises a region that is fully complementary to a target nucleic acid and is at least 80% complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain such embodiments, the region of full complementarity is from 6 to 14 nucleobases in length.
  • hybridization of an antisense compound results in recruitment of a protein that cleaves of the target nucleic acid.
  • certain antisense compounds result in RNase H mediated cleavage of target nucleic acid.
  • RNase H is a cellular endonuclease that cleaves the RNA strand of an RNA:DNA duplex.
  • the "DNA” in such an RNA:DNA duplex need not be unmodified DNA.
  • the invention provides antisense compounds that are sufficiently "DNA-like" to elicit RNase H activity.
  • DNA-like antisense compounds include, but are not limited to gapmers having unmodified deoxyfuronose sugar moieties in the nucleosides of the gap and modified sugar moieties in the nucleosides of the wings.
  • Antisense activities may be observed directly or indirectly.
  • observation or detection of an antisense activity involves observation or detection of a change in an amount of a target nucleic acid or protein encoded by such target nucleic acid; a change in the ratio of splice variants of a nucleic acid or protein; and/or a phenotypic change in a cell or animal.
  • compounds comprising oligonucleotides having a gapmer nucleoside motif described herein have desirable properties compared to non-gapmer oligonucleotides or to gapmers having other motifs. In certain circumstances, it is desirable to identify motifs resulting in a favorable combination of potent antisense activity and relatively low toxicity. In certain embodiments, compounds of the present invention have a favorable therapeutic index (measure of activity divided by measure of toxicity).
  • antisense compounds provided are selective for a target relative to a non-target nucleic acid.
  • the nucleobase sequences of the target and non-target nucleic acids differ by no more than 4 differentiating nucleobases in the targeted region.
  • the nucleobase sequences of the target and non-target nucleic acids differ by no more than 3 differentiating nucleobases in the targeted region.
  • the nucleobase sequences of the target and non-target nucleic acids differ by no more than 2 differentiating nucleobases in the targeted region.
  • the nucleobase sequences of the target and non-target nucleic acids differ by a single differentiating nucleobase in the targeted region.
  • the target and non-target nucleic acids are transcripts from different genes.
  • the target and non-target nucleic acids are different alleles for the same gene.
  • the introduction of a mismatch between an antisense compound and a non-target nucleic acid may alter the RNase H cleavage site of a target nucleic acid compared to a non-target nucleic acid.
  • the target and non-target nucleic acids are not functionally related to one another (e.g., are transcripts from different genes).
  • the target and not-target nucleic acids are allelic variants of one another.
  • the allelic variant contains a single nucleotide polymorphism (SNP).
  • SNP single nucleotide polymorphism
  • a SNP is associated with a mutant allele.
  • a mutant SNP is associated with a disease.
  • a mutant SNP is associated with a disease, but is not causative of the disease.
  • mRNA and protein expression of a mutant allele is associated with disease.
  • antisense compounds are achieved, principally, by nucleobase complementarity. For example, if an antisense compound has no mismatches for a target nucleic acid and one or more mismatches for a non-target nucleic acid, some amount of selectivity for the target nucleic acid will result. In certain embodiments, provided herein are antisense compounds with enhanced selectivity (i.e. the ratio of activity for the target to the activity for non-target is greater).
  • a selective nucleoside comprises a particular feature or combination of features (e.g., chemical modification, motif, placement of selective nucleoside, and/or self-complementary region) that increases selectivity of an antisense compound compared to an antisense compound not having that feature or combination of features.
  • a feature or combination of features increases antisense activity for the target.
  • such feature or combination of features decreases activity for the target, but decreases activity for the non-target by a greater amount, thus resulting in an increase in selectivity.
  • a selective antisense compound comprises a modified nucleoside at that same position as a differentiating nucleobase (i.e., the selective nucleoside is modified). That modification may increase the difference in binding affinity of the antisense compound for the target relative to the non-target.
  • the chemical modification may increase the difference in RNAse H activity for the duplex formed by the antisense compound and its target compared to the RNase activity for the duplex formed by the antisense compound and the non-target.
  • the modification may exaggerate a structure that is less compatible for RNase H to bind, cleave and/or release the non-target.
  • an antisense compound binds its intended target to form a target duplex.
  • RNase H cleaves the target nucleic acid of the target duplex.
  • the same antisence compound hybridizes to a non-target to form a non-target duplex.
  • the non-target differs from the target by a single nucleobase within the target region, and so the antisense compound hybridizes with a single mismatch. Because of the mismatch, in certain embodiments, RNase H cleavage of the non-target may be reduced compared to cleavage of the target, but still occurs. In certain embodiments, though, the primary site of that cleavage of the non-target nucleic acid (primary non-target cleavage site) is different from that of the target. That is; the primary site is shifted due to the mismatch. In such a circumstance, one may use a modification placed in the antisense compound to disrupt RNase H cleavage at the primary non-target cleavage site. Such modification will result in reduced cleavage of the non-target, but will result little or no decrease in cleavage of the target. In certain embodiments, the modification is a modified sugar, nucleobase and/or linkage.
  • the primary non-target cleavage site is towards the 5'-end of the antisense compound, and the 5'-end of an antisense compound may be modified to prevent RNaseH cleavage.
  • the 5'-end of an antisense compound may be modified to prevent RNaseH cleavage.
  • one having skill in the art may modify the 5'-end of an antisense compound, or modify the nucleosides in the gap region of the 5'-end of the antisense compound, or modify the the 3'-most 5'-region nucleosides of the antisense compound to selectively inhibit RNaseH cleavage of the non-target nucleic acid duplex while retaining RNase H cleavage of the target nucleic acid duplex.
  • 1-3 of the 3'-most 5'-region nucleosides of the antisense compound comprises a bicyclic sugar moiety.
  • the target nucleic acid may have an allelic variant, e.g. a non-target nucleic acid, containing a single nucleotide polymorphism.
  • An antisense compound may be designed having a single nucleobase mismatch from the non-target nucleic acid, but which has full complementarity to the target nucleic acid. The mismatch between the antisense compound and the non-target nucleic acid may destabilize the antisense compound non-target nucleic acid duplex, and consequently the cleavage site of RNaseH may shift upstream towards the 5'-end of the antisense compound.
  • Modification of the 5'-end of the antisense compound or the gap region near the 5'-end of the antisense compound, or one or more of the 3'-most nucleosides of the 5'-wing region, will then prevent RNaseH cleavage of the non-target nucleic acid. Since the target nucleic acid is fully complementary to the antisense compound, the antisense compound and the target nucleic acid will form a more stabilized antisense compound-target nucleic acid duplex and the cleavage site of RnaseH will be more downstream, towards the 3' end of the antisense compound.
  • one or more of the 3'-most nucleosides of the 5'-wing region comprises a bicyclic sugar moiety. In certain embodiments, one or more of the 3'-most nucleosides of the 5'-wing region comprises a bicyclic sugar moiety selected from cEt and LNA. In certain embodiments, one or more of the 3'-most nucleosides of the 5'-wing region comprises cEt. In certain embodiments, one or more of the 3'-most nucleosides of the 5'-wing region comprises LNA.
  • the introduction of a mismatch between an antisense compound and a target nucleic acid may alter the RNase H cleavage site of a target nucleic acid compared to a non-target nucleic acid by shifting the RNaseH cleavage site downstream from the mismatch site and towards the 3'-end of the antisense compound.
  • the cleavage site of a target nucleic acid compared to a non-target nucleic acid has shifted downstream towards the 3'-end of the antisense compound
  • the 3'-end of an antisense compound may be modified to prevent RNaseH cleavage.
  • the target nucleic acid may have an allelic variant, e.g. a non-target nucleic acid, containing a single nucleotide polymorphism.
  • An antisense compound may be designed having a single nucleobase mismatch from the non-target nucleic acid, but which has full complementarity to target nucleic acid. The mismatch between the antisense compound and the non-target nucleic acid may destabilize the antisense compound-non-target nucleic acid duplex, and consequently the cleavage site of RNaseH may shift downstream towards the 3'-end of the antisense compound.
  • Modification of the 3'-end of the antisense compound, or one or more of the the 5'-most nucleosides of the 3'-wing region, or the gap region of the antisense compound near the 3'-end will then prevent RNaseH cleavage of the non-target nucleic acid. Since the target nucleic acid is fully complementary to the antisense compound, the antisense compound and the target nucleic acid will form a more stabilized antisense compound-target nucleic acid duplex and the cleavage site of RnaseH will be more upstream, towards the 5' end of the antisense compound.
  • one or more of the 5'-most nucleosides of the 3'-wing region comprises a bicyclic sugar moiety. In certain embodiments, one or more of the 5'-most nucleosides of the 3'-wing region comprises a bicyclic sugar moiety selected from cEt and LNA. In certain embodiments, one or more of the 5'-most nucleosides of the 3'-wing region comprises cEt. In certain embodiments, one or more of the 5'-most nucleosides of the 3'-wing region comprises LNA.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of one or more bicyclic nucleosides at the 3'-most 5'-wing nucleoside. In certain embodiments, the selectivity of antisense compounds having certain gaps, e.g. gaps of 7 nucleosides or longer, may be improved by the addition of two or more bicyclic nucleosides at the 3'-most 5'-wing nucleoside. In certain embodiments, the selectivity of antisense compounds having certain gaps, e.g.
  • gaps of 7 nucleosides or longer may be improved by the addition of one bicyclic nucleoside at the 3'-most 5'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of two bicyclic nucleosides at the 3'-most 5'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of three bicyclic nucleosides at the 3'-most 5'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps e.g.
  • the bicyclic nucleosides at the 3'-most 5'-wing nucleoside comprise cEt. In certain embodiments discussed above, the bicyclic nucleosides at the 3'-most 5'-wing nucleoside comprise LNA.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of one or more bicyclic nucleosides at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of two or more bicyclic nucleosides at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of one bicyclic nucleoside at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of two bicyclic nucleosides at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of three bicyclic nucleosides at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of four bicyclic nucleosides at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of four bicyclic nucleosides at the 3'-most 5'-wing nucleoside and the addition of one or more bicylic nucleosides at the 5'-most 3'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of one or more bicyclic nucleosides at the 3'-most 5'-wing nucleoside. In certain embodiments, the selectivity of antisense compounds having certain gaps, e.g. gaps of 7 nucleosides or shorter, may be improved by the addition of two or more bicyclic nucleosides at the 3'-most 5'-wing nucleoside. In certain embodiments, the selectivity of antisense compounds having certain gaps, e.g.
  • gaps of 7 nucleosides or shorter may be improved by the addition of one bicyclic nucleoside at the 3'-most 5'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of two bicyclic nucleosides at the 3'-most 5'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps may be improved by the addition of three bicyclic nucleosides at the 3'-most 5'-wing nucleoside.
  • the selectivity of antisense compounds having certain gaps e.g.
  • the bicyclic nucleosides at the 3'-most 5'-wing nucleoside comprise cEt. In certain embodiments discussed above, the bicyclic nucleosides at the 3'-most 5'-wing nucleoside comprise LNA.
  • Antisense compounds having certain specified motifs have enhanced selectivity, including, but not limited to motifs described above.
  • enhanced selectivity is achieved by oligonucleotides comprising any one or more of:
  • a target region and a region of a non-target nucleic acid differ by 1-4 differentiating nucleobase.
  • selective antisense compounds have a nucleobase sequence that aligns with the non-target nucleic acid with 1-4 mismatches.
  • a nucleoside of the antisense compound that corresponds to a differentiating nucleobase of the target nucleic acid is referred to herein as a target-selective nucleoside.
  • selective antisense compounds having a gapmer motif align with a non-target nucleic acid, such that a target-selective nucleoside is positioned in the gap.
  • a target-selective nucleoside is the 6 rd nucleoside of the gap from the 5'-end. In certain embodiments, a target-selective nucleoside is the 8 th nucleoside of the gap from the 3'-end. In certain embodiments, a target-selective nucleoside is the 7 th nucleoside of the gap from the 3'-end. In certain embodiments, a target-selective nucleoside is the 6 th nucleoside of the gap from the 3'-end. In certain embodiments, a target-selective nucleoside is the 5 th nucleoside of the gap from the 3' -end.
  • a target-selective nucleoside is the 4 th nucleoside of the gap from the 3'-end. In certain embodiments, a target-selective nucleoside is the 3 rd nucleoside of the gap from the 3'-end. In certain embodiments, a target-selective nucleoside is the 2 nd nucleoside of the gap from the 3'-end.
  • a target-selective nucleoside comprises a modified nucleoside. In certain embodiments, a target-selective nucleoside comprises a modified sugar. In certain embodiments, a target-selective nucleoside comprises a sugar surrogate. In certain embodiments, a target-selective nucleoside comprises a sugar surrogate selected from among HNA and F-HNA. In certain embodiments, a target-selective nucleoside comprises a 2'-substituted sugar moiety. In certain embodiments, a target-selective nucleoside comprises a 2'-substituted sugar moiety selected from among MOE, F and (ara)-F.
  • a target-selective nucleoside comprises a 5'-substituted sugar moiety. In certain embodiments, a target-selective nucleoside comprises a 5'-substituted sugar moiety selected from 5'- (R) -Me DNA. In certain embodiments, a target-selective nucleoside comprises a bicyclic sugar moiety. In certain embodiments, a target-selective nucleoside comprises a bicyclic sugar moiety selected from among cEt, and ⁇ -L-LNA. In certain embodiments, a target-selective nucleoside comprises a modified nucleobase. In certain embodiments, a target-selective nucleoside comprises a modified nucleobase selected from among 2-thiothymidine and 5-propyne uridine.
  • selective antisense compounds comprise one or more mismatched nucleobases relative to the target nucleic acid.
  • antisense activity against the target is reduced by such mismatch, but activity against the non-target is reduced by a greater amount.
  • selectivity is improved.
  • Any nucleobase other than the differentiating nucleobase is suitable for a mismatch.
  • the mismatch is specifically positioned within the gap of an oligonucleotide having a gapmer motif.
  • a mismatch relative to the target nucleic acid is at positions 1, 2, 3, 4, 5, 6, 7, or 8 from the 5'-end of the gap region.
  • a mismatch relative to the target nucleic acid is at positions 9, 8, 7, 6, 5, 4, 3, 2, 1 of the antisense compounds from the 3'-end of the gap region. In certain embodiments, a mismatch relative to the target nucleid acid is at positions 1, 2, 3, or 4 of the antisense compounds from the 5'-end of the wing region. In certain embodiments, a mismatch relative to the target nucleid acid is at positions 4, 3, 2, or 1 of the antisense compounds from the 3'-end of the wing region.
  • selective antisense compounds comprise a region that is not complementary to the target. In certain embodiments, such region is complementary to another region of the antisense compound. Such regions are referred to herein as self-complementary regions.
  • an antisense compound has a first region at one end that is complementary to a second region at the other end. In certain embodiments, one of the first and second regions is complementary to the target nucleic acid. Unless the target nucleic acid also includes a self-complementary region, the other of the first and second region of the antisense compound will not be complementary to the target nucleic acid.
  • certain antisense compounds have the following nucleobase motif:
  • such antisense compounds are expected to form self-structure, which is disrupted upon contact with a target nucleic acid.
  • Contact with a non-target nucleic acid is expected to disrupt the self-structure to a lesser degree, thus increasing selectivity compared to the same antisense compound lacking the self-complementary regions.
  • a single antisense compound may include any one, two, three, or more of: self-complementary regions, a mismatch relative to the target nucleic acid, a short nucleoside gap, an interrupted gap, and specific placement of the selective nucleoside.
  • an antisense compound of interest may modulate the expression of a target nucleic acid but possess undesirable properties.
  • an antisense compound of interest may have an undesirably high affinity for one or more non-target nucleic acids.
  • an antisense compound of interest may produce undesirable increases in ALT and/or AST levels when administered to an animal.
  • such an antisense compound of interest may produce undesirable increases in organ weight.
  • an antisense compound of interest effectively modulates the expression of a target nucleic acid, but possess one or more undesirable properties
  • a person having skill in the art may selectively incorporate one or more modifications into the antisense compound of interest that retain some or all of the desired property of effective modulation of expression of a target nucleic acid while reducing one or more of the antisense compound's undesirable properties.
  • the present invention provides methods of altering such an antisense compound of interest to form an improved antisense compound.
  • altering the number of nucleosides in the 5'-region, the 3'-region, and/or the central region of such an antisense compound of interest results in improved properties.
  • an antisense compound having a modification motif of 3-10-3 could be altered to result in an improved antisense compound having a modification motif of 4-9-3 or 5-8-3.
  • the modification state of one or more of nucleosides at or near the 3'-end of the central region may likewise be altered.
  • the modification of one or more of the nucleosides at or near the 5'-end and the 3'-end of the central region may be altered.
  • the central region becomes shorter relative to the central region of the original antisense compound of interest.
  • the modifications to the one or more nucleosides that had been part of the central region are the same as one or more modification that had been present in the 5'-region and/or the 3'-region of the original antisense compound of interest.
  • the improved antisense compound having a shortened central region may retain its ability to effectively modulate the expression of a target nucleic acid, but not possess some or all of the undesirable properties possessed by antisense compound of interest having a longer central region.
  • reducing the length of the central region reduces affinity for off-target nucleic acids.
  • reducing the length of the central region results in reduced cleavage of non-target nucleic acids by RNase H.
  • reducing the length of the central region does not produce undesirable increases in ALT levels.
  • reducing the length of the central region does not produce undesirable increases in AST levels.
  • reducing the length of the central region does not produce undesirable increases organ weights.
  • nucleobase sequence and overall length of an antisense compound it is possible to retain the same nucleobase sequence and overall length of an antisense compound while decreasing the length of the central region. In certain embodiments retaining the same nucleobase sequence and overall length of an antisense compound while decreasing the length of the central region ameliorates one or more undesirable properties of an antisense compound. In certain embodiments retaining the same nucleobase sequence and overall length of an antisense compound while decreasing the length of the central region ameliorates one or more undesirable properties of an antisense compound but does not substantially affect the ability of the antisense compound to modulate expression of a target nucleic acid. In certain such embodiments, two or more antisense compounds would have the same overall length and nucleobase sequence, but would have a different central region length, and different properties.
  • the length of the central region is 9 nucleobases. In certain embodiments, the length of the central region is 8 nucleobases. In certain embodiments, the length of the central region is 7 nucleobases. In certain embodiments, the central region consists of unmodified deoxynucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 5'-region, the 3'-region, or both the 5'-region and the 3'-region.
  • the length of the central region can be decreased by increasing the length of the 5'-region with modified nucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 5'-region with modified nucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 5'-region with modified nucleosides comprising a bicyclic sugar moiety selected from among: cEt, cMOE, LNA, ⁇ -LNA, ENA and 2'-thio LNA. In certain embodiments, the length of the central region can be decreased by increasing the length of the 5'-region with a cEt substituted sugar moiety.
  • the length of the central region can be decreased by increasing the length of the 5'-region with modified nucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 5'-region with modified nucleosides.
  • the length of the central region can be decreased by increasing the length of the 5'-region with 2'- O(CH 2 ) 2 -OCH 3 (MOE) substituted sugar moiety
  • the length of the central region can be decreased by increasing the length of the 3'-region with modified nucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 3'-region with modified nucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 3'-region with modified nucleosides comprising a bicyclic sugar moiety selected from among: cEt, cMOE, LNA, ⁇ -LNA, ENA and 2'-thio LNA. In certain embodiments, the length of the central region can be decreased by increasing the length of the 3'-region with a cEt substituted sugar moiety.
  • the length of the central region can be decreased by increasing the length of the 3'-region with modified nucleosides. In certain embodiments, the length of the central region can be decreased by increasing the length of the 3'-region with modified nucleosides.
  • the length of the central region can be decreased by increasing the length of the 3'-region with 2'- O(CH 2 ) 2 -OCH 3 (MOE) substituted sugar moiety
  • the length of the central region can be decreased by increasing the length of the 5'-region with modified nucleosides and increasing the length of the 3'-region with modified nucleosides.
  • antisense compounds comprise or consist of an oligonucleotide comprising a region that is complementary to a target nucleic acid.
  • the target nucleic acid is an endogenous RNA molecule.
  • the target nucleic acid is a non-coding RNA.
  • the target non-coding RNA is selected from: a long-non-coding RNA, a short non-coding RNA, an intronic RNA molecule, a snoRNA, a scaRNA, a microRNA (including pre-microRNA and mature microRNA), a ribosomal RNA, and promoter directed RNA.
  • the target nucleic acid encodes a protein.
  • the target nucleic acid is selected from: an mRNA and a pre-mRNA, including intronic, exonic and untranslated regions.
  • oligomeric compounds are at least partially complementary to more than one target nucleic acid.
  • antisense compounds of the present invention may mimic microRNAs, which typically bind to multiple targets.
  • the target nucleic acid is a nucleic acid other than a mature mRNA. In certain embodiments, the target nucleic acid is a nucleic acid other than a mature mRNA or a microRNA. In certain embodiments, the target nucleic acid is a non-coding RNA other than a microRNA. In certain embodiments, the target nucleic acid is a non-coding RNA other than a microRNA or an intronic region of a pre-mRNA. In certain embodiments, the target nucleic acid is a long non-coding RNA. In certain embodiments, the target RNA is an mRNA. In certain embodiments, the target nucleic acid is a pre-mRNA.
  • the target region is entirely within an intron. In certain embodiments, the target region spans an intron/exon junction. In certain embodiments, the target region is at least 50% within an intron. In certain embodiments, the target nucleic acid is selected from among non-coding RNA, including exonic regions of pre-mRNA. In certain embodiments, the target nucleic acid is a ribosomal RNA (rRNA). In certain embodiments, the target nucleic acid is a non-coding RNA associated with splicing of other pre-mRNAs. In certain embodiments, the target nucleic acid is a nuclear-retained non-coding RNA.
  • rRNA ribosomal RNA
  • antisense compounds described herein are complementary to a target nucleic acid comprising a single-nucleotide polymorphism.
  • the antisense compound is capable of modulating expression of one allele of the single-nucleotide polymorphism-containing-target nucleic acid to a greater or lesser extent than it modulates another allele.
  • an antisense compound hybridizes to a single-nucleotide polymorphism-containing-target nucleic acid at the single-nucleotide polymorphism site.
  • the target nucleic acid is a Huntingtin gene transcript.
  • the target nucleic acid is a single-nucleotide polymorphism-containing-target nucleic acid of a Huntingtin gene transcript. In certain embodiments, the target nucleic acid is not a Huntingtin gene transcript. In certain embodiments, the target nucleic acid is a single-nucleotide polymorphism-containing-target nucleic acid of a gene transcript other than Huntingtin. In certain embodiments, the target nucleic acid is any nucleic acid other than a Huntingtin gene transcript.
  • the invention provides selective antisense compounds that have greater activity for a target nucleic acid than for a homologous or partially homologous non-target nucleic acid.
  • the target and non-target nucleic acids are not functionally related to one another (e.g., are transcripts from different genes).
  • the target and not-targe nucleic acids are allelic variants of one another.
  • Certain embodiments of the present invention provide methods, compounds, and compositions for selectively inhibiting mRNA and protein expression of an allelic variant of a particular gene or DNA sequence.
  • the allelic variant contains a single nucleotide polymorphism (SNP).
  • SNP single nucleotide polymorphism
  • a mutant SNP is associated with a disease. In certain embodiments a mutant SNP is associated with a disease, but is not causative of the disease. In certain embodiments, mRNA and protein expression of a mutant allele is associated with disease.
  • the expressed gene product of a mutant allele results in aggregation of the mutant proteins causing disease. In certain embodiments, the expressed gene product of a mutant allele results in gain of function causing disease.
  • genes with an autosomal dominant mutation resulting in a toxic gain of function of the protein are the APP gene encoding amyloid precursor protein involved in Alzheimer's disease ( Gene, 371: 68, 2006 ); the PrP gene encoding prion protein involved in Creutzfeldt-Jakob disease and in fatal familial insomnia ( Nat. Med. 1997, 3: 1009 ); GFAP gene encoding glial fibrillary acidic protein involved in Alexander disease ( J. Neurosci.
  • alpha-synuclein gene encoding alpha-synuclein protein involved in Parkinson's disease ( J. Clin. Invest. 2003, 111: 145 ); SOD-1 gene encoding the SOD-1 protein involved in amyotrophic lateral sclerosis ( Science 1998, 281: 1851 ); atrophin-1 gene encoding atrophin-1 protein involved in dentato-rubral and pallido-luysian atrophy (DRPA) ( Trends Mol. Med. 2001, 7: 479 ); SCA1 gene encoding ataxin-1 protein involved in spino-cerebellar ataxia-1 (SCA1) ( Protein Sci.
  • Ltk gene encoding leukocyte tyrosine kinase protein involved in systemic lupus erythematosus ( Hum. Mol. Gen. 2004, 13: 171 ); PCSK9 gene encoding PCSK9 protein involved in hypercholesterolemia ( Hum Mutat. 2009, 30: 520 ); prolactin receptor gene encoding prolactin receptor protein involved in breast tumors ( Proc. Natl. Assoc. Sci. 2008, 105: 4533 ); CCL5 gene encoding the chemokine CCL5 involved in COPD and asthma ( Eur. Respir. J.
  • PTPN22 gene encoding PTPN22 protein involved in Type 1 diabetes, Rheumatoid arthritis, Graves disease, and SLE ( Proc. Natl. Assoc. Sci. 2007, 104: 19767 ); androgen receptor gene encoding the androgen receptor protein involved in spinal and bulbar muscular atrophy or Kennedy's disease ( J Steroid Biochem. Mol. Biol. 2008, 108: 245 ); CHMP4B gene encoding chromatin modifying protein-4B involved in progressive childhood posterior subcapsular cataracts ( Am. J. Hum.
  • AVP gene encoding arginine vasopressin protein in stress-related disorders such as anxiety disorders and comorbid depression ( CNS Neurol. Disord. Drug Targets 2006, 5: 167 ); GNAS gene encoding G proteins involved in congenital visual defects, hypertension, metabolic syndrome ( Trends Pharmacol. Sci. 2006, 27: 260 ); APAF1 gene encoding APAF1 protein involved in a predisposition to major depression ( Mol. Psychiatry 2006, 11: 76 ); TGF-beta1 gene encoding TGF-beta1 protein involved in breast cancer and prostate cancer ( Cancer Epidemiol. Biomarkers Prev.
  • AChR gene encoding acetylcholine receptor involved in congential myasthenic syndrome ( Neurology 2004, 62: 1090 ); P2Y12 gene encoding adenosine diphosphate (ADP) receptor protein involved in risk of peripheral arterial disease ( Circulation 2003, 108: 2971 ); LQT1 gene encoding LQT1 protein involved in atrial fibrillation ( Cardiology 2003, 100: 109 ); RET protooncogene encoding RET protein involved in sporadic pheochromocytoma ( J. Clin. Endocrinol. Metab.
  • CA4 gene encoding carbonic anhydrase 4 protein, CRX gene encoding cone-rod homeobox transcription factor protein, FSCN2 gene encoding retinal fascin homolog 2 protein, IMPDH1 gene encoding inosine monophosphate dehydrogenase 1 protein, NR2E3 gene encoding nuclear receptor subfamily 2 group E3 protein, NRL gene encoding neural retina leucine zipper protein, PRPF3 (RP18) gene encoding pre-mRNA splicing factor 3 protein, PRPF8 (RP13) gene encoding pre-mRNA splicing factor 8 protein, PRPF31 (RP11) gene encoding pre-mRNA splicing factor 31 protein, RDS gene encoding peripherin 2 protein, ROM1 gene encoding rod outer membrane protein 1 protein, RHO gene encoding rhodopsin protein, RP1 gene encoding RP1 protein, RPGR gene encoding retinitis pigmentosa G
  • the mutant allele is associated with any disease from the group consisting of Alzheimer's disease, Creutzfeldt-Jakob disease, fatal familial insomnia, Alexander disease, Parkinson's disease, amyotrophic lateral sclerosis, dentato-rubral and pallido-luysian atrophy DRPA, spino-cerebellar ataxia, Torsion dystonia, cardiomyopathy, chronic obstructive pulmonary disease (COPD), liver disease, hepatocellular carcinoma, systemic lupus erythematosus, hypercholesterolemia, breast cancer, asthma, Type 1 diabetes, Rheumatoid arthritis, Graves disease, SLE, spinal and bulbar muscular atrophy, Kennedy's disease, progressive childhood posterior subcapsular cataracts, cholesterol gallstone disease, arthrosclerosis, cardiovascular disease, primary hypercalciuria, alpha-thallasemia, obsessive compulsive disorder, Anxiety, comorbid depression, congenital visual defects, hypertension, metabolic syndrome,
  • any disease
  • an allelic variant of huntingtin is selectively reduced.
  • Nucleotide sequences that encode huntingtin include, without limitation, the following: GENBANK Accession No. NT_006081.18, truncated from nucleotides 1566000 to 1768000 (replaced by GENBANK Accession No. NT_006051), incorporated herein as SEQ ID NO: 1, and NM_002111.6, incorporated herein as SEQ ID NO: 2.
  • Table 14 provides SNPs found in the GM04022, GM04281, GM02171, and GM02173B cell lines. Also provided are the allelic variants found at each SNP position, the genotype for each of the cell lines, and the percentage of HD patients having a particular allelic variant. For example, the two allelic variants for SNP rs6446723 are T and C.
  • the GM04022 cell line is heterozygous TC
  • the GM02171 cell line is homozygous CC
  • the GM02173 cell line is heterozygous TC
  • the GM04281 cell line is homozygous TT.
  • Fifty percent of HD patients have a T at SNP position rs6446723.
  • provided herein are methods of treating an animal or individual comprising administering one or more pharmaceutical compositions as described herein.
  • the individual or animal has Huntington's disease.
  • compounds targeted to huntingtin as described herein may be administered to reduce the severity of physiological symptoms of Huntington's disease. In certain embodiments, compounds targeted to huntingtin as described herein may be administered to reduce the rate of degeneration in an individual or an animal having Huntington's disease. In certain embodiments, compounds targeted to huntingtin as described herein may be administered regeneration function in an individual or an animal having Huntington's disease. In certain embodiments, symptoms of Huntingtin's disease may be reversed by treatment with a compound as described herein.
  • compounds targeted to huntingtin as described herein may be administered to ameliorate one or more symptoms of Huntington's disease.
  • administration of compounds targeted to huntingtin as described herein may improve the symptoms of Huntington's disease as measured by any metric known to those having skill in the art.
  • administration of compounds targeted to huntingtin as described herein may improve a rodent's rotaraod assay performance.
  • administration of compounds targeted to huntingtin as described herein may improve a rodent's plus maze assay.
  • administration of compounds targeted to huntingtin as described herein may improve a rodent's open field assay performance.
  • provided herein are methods for ameliorating a symptom associated with Huntington's disease in a subject in need thereof.
  • a method for reducing the rate of onset of a symptom associated with Huntington's disease In certain embodiments, provided is a method for reducing the severity of a symptom associated with Huntington's disease. In certain embodiments, provided is a method for regenerating neurological function as shown by an improvement of a symptom associated with Huntington's disease.
  • the methods comprise administering to an individual or animal in need thereof a therapeutically effective amount of a compound targeted to a huntingtin nucleic acid.
  • Huntington's disease is characterized by numerous physical, neurological, psychiatric, and/or peripheral symptoms. Any symptom known to one of skill in the art to be associated with Huntington's disease can be ameliorated or otherwise modulated as set forth above in the methods described above.
  • the symptom is a physical symptom selected from the group consisting of restlessness, lack of coordination, unintentionally initiated motions, unintentionally uncompleted motions, unsteady gait, chorea, rigidity, writhing motions, abnormal posturing, instability, abnormal facial expressions, difficulty chewing, difficulty swallowing, difficulty speaking, seizure, and sleep disturbances.
  • the symptom is a cognitive symptom selected from the group consisting of impaired planning, impaired flexibility, impaired abstract thinking, impaired rule acquisition, impaired initiation of appropriate actions, impaired inhibition of inappropriate actions, impaired short-term memory, impaired long-term memory, paranoia, disorientation, confusion, hallucination and dementia.
  • the symptom is a psychiatric symptom selected from the group consisting of anxiety, depression, blunted affect, egocentrisms, aggression, compulsive behavior, irritability and suicidal ideation.
  • the symptom is a peripheral symptom selected from the group consisting of reduced brain mass, muscle atrophy, cardiac failure, impaired glucose tolerance, weight loss, osteoporosis, and testicular atrophy.
  • the symptom is restlessness. In certain embodiments, the symptom is lack of coordination. In certain embodiments, the symptom is unintentionally initiated motions. In certain embodiments, the symptom is unintentionally uncompleted motions. In certain embodiments, the symptom is unsteady gait. In certain embodiments, the symptom is chorea. In certain embodiments, the symptom is rigidity. In certain embodiments, the symptom is writhing motions. In certain embodiments, the symptom is abnormal posturing. In certain embodiments, the symptom is instability. In certain embodiments, the symptom is abnormal facial expressions. In certain embodiments, the symptom is difficulty chewing. In certain embodiments, the symptom is difficulty swallowing. In certain embodiments, the symptom is difficulty speaking. In certain embodiments, the symptom is seizures. In certain embodiments, the symptom is sleep disturbances.
  • the symptom is impaired planning. In certain embodiments, the symptom is impaired flexibility. In certain embodiments, the symptom is impaired abstract thinking. In certain embodiments, the symptom is impaired rule acquisition. In certain embodiments, the symptom is impaired initiation of appropriate actions. In certain embodiments, the symptom is impaired inhibition of inappropriate actions. In certain embodiments, the symptom is impaired short-term memory. In certain embodiments, the symptom is impaired long-term memory. In certain embodiments, the symptom is paranoia. In certain embodiments, the symptom is disorientation. In certain embodiments, the symptom is confusion. In certain embodiments, the symptom is hallucination. In certain embodiments, the symptom is dementia.
  • the symptom is anxiety. In certain embodiments, the symptom is depression. In certain embodiments, the symptom is blunted affect. In certain embodiments, the symptom is egocentrism. In certain embodiments, the symptom is aggression. In certain embodiments, the symptom is compulsive behavior. In certain embodiments, the symptom is irritability. In certain embodiments, the symptom is suicidal ideation.
  • the symptom is reduced brain mass. In certain embodiments, the symptom is muscle atrophy. In certain embodiments, the symptom is cardiac failure. In certain embodiments, the symptom is impaired glucose tolerance. In certain embodiments, the symptom is weight loss. In certain embodiments, the symptom is osteoporosis. In certain embodiments, the symptom is testicular atrophy.
  • symptoms of Huntington's disease may be quantifiable.
  • osteoporosis may be measured and quantified by, for example, bone density scans.
  • the symptom may be reduced by about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.
  • provided are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein.
  • the individual has Huntington's disease.
  • administering results in reduction of huntingtin expression by at least about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.
  • compositions comprising an antisense compound targeted to huntingtin are used for the preparation of a medicament for treating a patient suffering or susceptible to Huntington's disease.
  • the present invention provides pharmaceutical compositions comprising one or more antisense compound.
  • such pharmaceutical composition comprises a suitable pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition comprises a sterile saline solution and one or more antisense compound.
  • such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound.
  • the sterile saline is pharmaceutical grade saline.
  • a pharmaceutical composition comprises one or more antisense compound and sterile water.
  • a pharmaceutical composition consists of one or more antisense compound and sterile water.
  • the sterile saline is pharmaceutical grade water.
  • a pharmaceutical composition comprises one or more antisense compound and phosphate-buffered saline (PBS). In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile phosphate-buffered saline (PBS). In certain embodiments, the sterile saline is pharmaceutical grade PBS.
  • antisense compounds may be admixed with pharmaceutically acceptable active and/or inert substances for the preparation of pharmaceutical compositions or formulations.
  • Compositions and methods for the formulation of pharmaceutical compositions depend on a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.
  • compositions comprising antisense compounds encompass any pharmaceutically acceptable salts, esters, or salts of such esters.
  • pharmaceutical compositions comprising antisense compounds comprise one or more oligonucleotide which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof.
  • the disclosure is also drawn to pharmaceutically acceptable salts of antisense compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.
  • a prodrug can include the incorporation of additional nucleosides at one or both ends of an oligomeric compound which are cleaved by endogenous nucleases within the body, to form the active antisense oligomeric compound.
  • Lipid moieties have been used in nucleic acid therapies in a variety of methods.
  • the nucleic acid is introduced into preformed liposomes or lipoplexes made of mixtures of cationic lipids and neutral lipids.
  • DNA complexes with mono- or poly-cationic lipids are formed without the presence of a neutral lipid.
  • a lipid moiety is selected to increase distribution of a pharmaceutical agent to a particular cell or tissue.
  • a lipid moiety is selected to increase distribution of a pharmaceutical agent to fat tissue.
  • a lipid moiety is selected to increase distribution of a pharmaceutical agent to muscle tissue.
  • compositions provided herein comprise one or more modified oligonucleotides and one or more excipients.
  • excipients are selected from water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylase, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose and polyvinylpyrrolidone.
  • a pharmaceutical composition provided herein comprises a delivery system.
  • delivery systems include, but are not limited to, liposomes and emulsions. Certain delivery systems are useful for preparing certain pharmaceutical compositions including those comprising hydrophobic compounds. In certain embodiments, certain organic solvents such as dimethylsulfoxide are used.
  • a pharmaceutical composition provided herein comprises one or more tissue-specific delivery molecules designed to deliver the one or more pharmaceutical agents of the present invention to specific tissues or cell types.
  • pharmaceutical compositions include liposomes coated with a tissue-specific antibody.
  • a pharmaceutical composition provided herein comprises a co-solvent system.
  • co-solvent systems comprise, for example, benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • co-solvent systems are used for hydrophobic compounds.
  • VPD co-solvent system is a solution of absolute ethanol comprising 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80 TM and 65% w/v polyethylene glycol 300.
  • co-solvent systems may be varied considerably without significantly altering their solubility and toxicity characteristics.
  • identity of co-solvent components may be varied: for example, other surfactants may be used instead of Polysorbate 80 TM ; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • a pharmaceutical composition provided herein is prepared for oral administration. In certain embodiments, pharmaceutical compositions are prepared for buccal administration.
  • a pharmaceutical composition is prepared for administration by injection (e.g., intravenous, subcutaneous, intramuscular, etc.).
  • a pharmaceutical composition comprises a carrier and is formulated in aqueous solution, such as water or physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • other ingredients are included (e.g., ingredients that aid in solubility or serve as preservatives).
  • injectable suspensions are prepared using appropriate liquid carriers, suspending agents and the like.
  • compositions for injection are suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • Certain solvents suitable for use in pharmaceutical compositions for injection include, but are not limited to, lipophilic solvents and fatty oils, such as sesame oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, and liposomes.
  • Aqueous injection suspensions may contain.
  • the compounds and compositions as described herein are administered parenterally.
  • parenteral administration is by infusion. Infusion can be chronic or continuous or short or intermittent. In certain embodiments, infused pharmaceutical agents are delivered with a pump. In certain embodiments, parenteral administration is by injection.
  • compounds and compositions are delivered to the CNS. In certain embodiments, compounds and compositions are delivered to the cerebrospinal fluid. In certain embodiments, compounds and compositions are administered to the brain parenchyma. In certain embodiments, compounds and compositions are delivered to an animal by intrathecal administration, or intracerebroventricular administration. Broad distribution of compounds and compositions, described herein, within the central nervous system may be achieved with intraparenchymal administration, intrathecal administration, or intracerebroventricular administration.
  • parenteral administration is by injection.
  • the injection may be delivered with a syringe or a pump.
  • the injection is a bolus injection.
  • the injection is administered directly to a tissue, such as striatum, caudate, cortex, hippocampus and cerebellum.
  • delivery of a compound or composition described herein can affect the pharmacokinetic profile of the compound or composition.
  • injection of a compound or composition described herein, to a targeted tissue improves the pharmacokinetic profile of the compound or composition as compared to infusion of the compound or composition.
  • the injection of a compound or composition improves potency compared to broad diffusion, requiring less of the compound or composition to achieve similar pharmacology.
  • similar pharmacology refers to the amount of time that a target mRNA and/or target protein is downregulated (e.g. duration of action).
  • methods of specifically localizing a pharmaceutical agent decreases median effective concentration (EC50) by a factor of about 50 (e.g. 50 fold less concentration in tissue is required to achieve the same or similar pharmacodynamic effect).
  • methods of specifically localizing a pharmaceutical agent decreases median effective concentration (EC50) by a factor of 20, 25, 30, 35, 40, 45 or 50.
  • the pharmaceutical agent in an antisense compound as further described herein.
  • the targeted tissue is brain tissue.
  • the targeted tissue is striatal tissue.
  • decreasing EC50 is desirable because it reduces the dose required to achieve a pharmacological result in a patient in need thereof.
  • an antisense oligonucleotide is delivered by injection or infusion once every month, every two months, every 90 days, every 3 months, every 6 months, twice a year or once a year.
  • one or more pharmaceutical compositions are co-administered with one or more other pharmaceutical agents.
  • such one or more other pharmaceutical agents are designed to treat the same disease, disorder, or condition as the one or more pharmaceutical compositions described herein.
  • such one or more other pharmaceutical agents are designed to treat a different disease, disorder, or condition as the one or more pharmaceutical compositions described herein.
  • such one or more other pharmaceutical agents are designed to treat an undesired side effect of one or more pharmaceutical compositions as described herein.
  • one or more pharmaceutical compositions are co-administered with another pharmaceutical agent to treat an undesired effect of that other pharmaceutical agent.
  • one or more pharmaceutical compositions are co-administered with another pharmaceutical agent to produce a combinational effect. In certain embodiments, one or more pharmaceutical compositions are co-administered with another pharmaceutical agent to produce a synergistic effect.
  • one or more pharmaceutical compositions and one or more other pharmaceutical agents are administered at the same time. In certain embodiments, one or more pharmaceutical compositions and one or more other pharmaceutical agents are administered at different times. In certain embodiments, one or more pharmaceutical compositions and one or more other pharmaceutical agents are prepared together in a single formulation. In certain embodiments, one or more pharmaceutical compositions and one or more other pharmaceutical agents are prepared separately.
  • pharmaceutical agents that may be co-administered with a pharmaceutical composition of include antipsychotic agents, such as, e.g., haloperidol, chlorpromazine, clozapine, quetapine, and olanzapine; antidepressant agents, such as, e.g., fluoxetine, sertraline hydrochloride, venlafaxine and nortriptyline; tranquilizing agents such as, e.g., benzodiazepines, clonazepam, paroxetine, venlafaxin, and beta-blockers; mood-stabilizing agents such as, e.g ., lithium, valproate, lamotrigine, and carbamazepine; paralytic agents such as, e.g ., Botulinum toxin; and/or other experimental agents including, but not limited to, tetrabenazine (Xenazine), creatine, conezyme Q10, trehalose, docosahex
  • RNA nucleoside comprising a 2'-OH sugar moiety and a thymine base
  • RNA methylated uracil
  • nucleic acid sequences provided herein are intended to encompass nucleic acids containing any combination of natural or modified RNA and/or DNA, including, but not limited to such nucleic acids having modified nucleobases.
  • an oligomeric compound having the nucleobase sequence "ATCGATCG” encompasses any oligomeric compounds having such nucleobase sequence, whether modified or unmodified, including, but not limited to, such compounds comprising RNA bases, such as those having sequence "AUCGAUCG” and those having some DNA bases and some RNA bases such as “AUCGATCG” and oligomeric compounds having other modified or naturally occurring bases, such as "AT me CGAUCG,” wherein me C indicates a cytosine base comprising a methyl group at the 5-position.
  • oligomeric compounds are assigned a "Sequence Code.” Oligomeric compounds having the same Sequence Code have the same nucleobase sequence. Oligomeric compounds having different Sequence Codes have different nucleobase sequences.
  • Example 1 Modified antisense oligonucleotides targeting human Target-X
  • Antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. ISIS 407939, which was described in an earlier publication ( WO 2009/061851 ) was also tested.
  • the newly designed chimeric antisense oligonucleotides and their motifs are described in Table 15.
  • Nucleosides followed by “d” indicate 2'-deoxyribonucleosides.
  • Nucleosides followed by “k” indicate 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) nucleosides.
  • Nucleosides followed by "e” indicate 2'-O-methoxyethyl (2'-MOE) nucleosides.
  • “N” indicates modified or naturally occurring nucleobases (A, T, C, G, U, or 5-methyl C).
  • Each gapmer listed in Table 15 is targeted to the human Target-X genomic sequence.
  • Results are presented as percent inhibition of Target-X, relative to untreated control cells, and indicate that several of the newly designed antisense oligonucleotides are more potent than ISIS 407939. A total of 771 oligonucleotides were tested. Only those oligonucleotides which were selected for further studies are shown in Table 15. Each of the newly designed antisense oligonucleotides provided in Table 1 achieved greater than 80% inhibition and, therefore, are more active than ISIS 407939.
  • Example 2 Modified antisense oligonucleotides comprising 6'-(S)-CH 3 bicyclic nucleoside (cEt) and F-HNA modifications targeting human Target-X
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. ISIS 407939 was also tested.
  • the chimeric antisense oligonucleotides and their motifs are described in Table 16.
  • Nucleosides followed by “d” indicate 2'-deoxyribonucleosides.
  • Nucleosides followed by “k” indicate 6'-(S)-CH 3 bicyclic nucleosides (e.g cEt).
  • Nucleosides followed by "e” indicate 2'-O-methoxyethyl (2'-MOE) modified nucleosides.
  • Nucleosides followed by 'g' indicate F-HNA modified nucleosides.
  • “N” indicates modified or naturally occurring nucleobases (A, T, C, G, U, or 5-methyl C).
  • Each gapmer listed in Table 16 is targeted to the human Target-X genomic sequence.
  • Results are presented as percent inhibition of Target-X, relative to untreated control cells, and demonstrate that several of the newly designed gapmers are more potent than ISIS 407939.
  • a total of 765 oligonucleotides were tested. Only those oligonucleotides which were selected for further studies are shown in Table 16. All but one of the newly designed antisense oligonucleotides provided in Table 16 achieved greater than 30% inhibition and, therefore, are more active than ISIS 407939.
  • Example 3 Modified antisense oligonucleotides comprising 2'-MOE and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X
  • the newly designed chimeric antisense oligonucleotides are 16 nucleotides in length and their motifs are described in Table 17.
  • the chemistry column of Table 17 presents the sugar motif of each oligonucleotide, wherein "e” indicates a 2'-O-methoxyethyl (2'-MOE) nucleoside, "k” indicates a 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) and "d” indicates a 2'-deoxyribonucleoside.
  • Each gapmer listed in Table 17 is targeted to the human Target-X genomic sequence.
  • ISIS 403052, ISIS 407594, ISIS 407606, ISIS 407939, and ISIS 416438 Cultured Hep3B cells at a density of 20,000 cells per well were transfected using electroporation with 2,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR. Human primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels. Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. Also tested were ISIS 403094, ISIS 407641, ISIS 407643, ISIS 407662, ISIS 407900, ISIS 407910, ISIS 407935, ISIS 407936, ISIS 407939, ISIS 416446, ISIS 416449, ISIS 416455, ISIS 416472, ISIS 416477, ISIS 416507, ISIS 416508, ISIS 422086, ISIS 422087, ISIS 422140, and ISIS 422142, 5-10-5 2'-MOE gapmers targeting human Target-X, which were described in an earlier publication ( WO 2009/061851 ), incorporated herein by reference..
  • Each gapmer listed in Table 18 is targeted to the human Target-X genomic sequence.
  • Human primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of Target-X, relative to untreated control cells. A total of 916 oligonucleotides were tested. Only those oligonucleotides which were selected for further studies are shown in Tables 18 and 19.
  • Example 5 Modified chimeric antisense oligonucleotides comprising 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications at 5' and 3' wing regions targeting human Target-X
  • ISIS 407939 which was described in an earlier publication ( WO 2009/061851 ) were also tested.
  • the newly designed chimeric antisense oligonucleotides in Table 20 were designed as 2-10-2 cEt gapmers.
  • the newly designed gapmers are 14 nucleosides in length, wherein the central gap segment comprises of ten 2'-deoxyribonucleosides and is flanked by wing segments on the 5' direction and the 3' direction comprising five nucleosides each.
  • Each nucleoside in the 5' wing segment and each nucleoside in the 3' wing segment comprises 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modification.
  • Each gapmer listed in Table 20 is targeted to the human Target-X genomic sequence.
  • Human primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of Target-X, relative to untreated control cells. A total of 614 oligonucleotides were tested.
  • Example 6 Modified chimeric antisense oligonucleotides comprising 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications at 5' and 3' wing regions targeting human Target-X
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. Also tested was ISIS 407939, a 5-10-5 MOE gapmer targeting human Target-X, which was described in an earlier publication ( WO 2009/061851 ). ISIS 472998 and ISIS 473046, described in the Examples above were also included in the screen.
  • the newly designed chimeric antisense oligonucleotides in Table 21 were designed as 2-10-2 cEt gapmers.
  • the newly designed gapmers are 14 nucleosides in length, wherein the central gap segment comprises of ten 2'-deoxyribonucleosides and is flanked by wing segments on the 5' direction and the 3' direction comprising five nucleosides each.
  • Each nucleoside in the 5' wing segment and each nucleoside in the 3' wing segment comprise 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modification.
  • Each gapmer listed in Table 21 is targeted to the human Target-X genomic sequence.
  • Human primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of Target-X, relative to untreated control cells. A total of 757 oligonucleotides were tested.
  • Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.67 ⁇ M, 2.00 ⁇ M, 1.11 ⁇ M, and 6.00 ⁇ M concentrations of antisense oligonucleotide, as specified in Table 22.
  • RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR.
  • Human Target-X primer probe set RTS2927 was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • IC 50 half maximal inhibitory concentration of each oligonucleotide is also presented in Table 22. As illustrated in Table 22, Target-X mRNA levels were reduced in a dose-dependent manner in antisense oligonucleotide treated cells. The data also confirms that several of the newly designed gapmers are more potent than ISIS 407939 of the previous publication.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells. As illustrated in Table 23, Target-X mRNA levels were reduced in a dose-dependent manner in antisense oligonucleotide treated cells. The data also confirms that several of the newly designed gapmers are more potent than ISIS 407939.
  • Example 9 Modified chimeric antisense oligonucleotides comprising 2'-methoxyethyl (2'-MOE) modifications at 5' and 3' wing regions targeting human Target-X
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. Also tested were ISIS 403052, ISIS 407939, ISIS 416446, ISIS 416472, ISIS 416507, ISIS 416508, ISIS 422087, ISIS 422096, ISIS 422130, and ISIS 422142 which were described in an earlier publication ( WO 2009/061851 ), incorporated herein by reference. ISIS 490149, ISIS 490197, ISIS 490209, ISIS 490275, ISIS 490277, and ISIS 490424, described in the Examples above, were also included in the screen.
  • the newly designed chimeric antisense oligonucleotides in Table 24 were designed as 3-10-4 2'-MOE gapmers. These gapmers are 17 nucleosides in length, wherein the central gap segment comprises of ten 2'-deoxyribonucleosides and is flanked by wing segments on the 5' direction with three nucleosides and the 3' direction with four nucleosides. Each nucleoside in the 5' wing segment and each nucleoside in the 3' wing segment has 2'-MOE modifications.
  • Each gapmer listed in Table 24 is targeted to the human Target-X genomic sequence.
  • oligonucleotides Activity of the newly designed oligonucleotides was compared to ISIS 403052, ISIS 407939, ISIS 416446, ISIS 416472, ISIS 416507, ISIS 416508, ISIS 422087, ISIS 422096, ISIS 422130, and ISIS 422142.
  • Cultured Hep3B cells at a density of 20,000 cells per well were transfected using electroporation with 2,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR. Human primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells. A total of 272 oligonucleotides were tested. Only those oligonucleotides which were selected for further studies are shown in Table 24. Several of the newly designed antisense oligonucleotides provided in Table 24 are more potent than antisense oligonucleotides from the previous publication.
  • IC 50 half maximal inhibitory concentration
  • RNA samples were plated at a density of 20,000 cells per well and transfected using electroporation with 0.3125 ⁇ M, 0.625 ⁇ M, 1.25 ⁇ M, 2.50 ⁇ M, 5.00 ⁇ M and 10.00 ⁇ M concentrations of antisense oligonucleotide, as specified in Table 26.
  • RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR.
  • Human Target-X primer probe set RTS2927 was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • Target-X mRNA levels were reduced in a dose-dependent manner in antisense oligonucleotide treated cells.
  • the data also confirms that the newly designed gapmers are more potent than gapmers from the previous publication.
  • Table 26 Dose-dependent antisense inhibition of human Target-X in Hep3B cells using electroporation ISIS No 0.3125 ⁇ M 0.625 ⁇ M 1.250 ⁇ M 2.500 ⁇ M 5.000 ⁇ M 10.000 ⁇ M IC 50 ( ⁇ M) 407935 30 49 75 86 91 94 0.6 407939 30 48 61 78 85 90 0.8 416446 27 52 63 75 85 90 0.7 416472 38 51 72 83 88 94 0.5 416507 58 81 76 84 89 92 ⁇ 0.3 416549 52 67 75 81 88 89 0.3 422086 48 49 68 78 86 91 0.5 422087 30 56 66 83 72 92 0.6 422096 47 63 70 77 83 85
  • Example 12 Tolerability of 2'-MOE gapmers targeting human Target-X in BALB/c mice
  • mice are a multipurpose mice model, frequently utilized for safety and efficacy testing.
  • the mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • mice Groups of male BALB/c mice were injected subcutaneously twice a week for 3 weeks with 50 mg/kg of ISIS 407935, ISIS 416472, ISIS 416549, ISIS 422086, ISIS 422087, ISIS 422096, ISIS 422142, ISIS 490103, ISIS 490149, ISIS 490196, ISIS 490208, ISIS 490209, ISIS 513419, ISIS 513420, ISIS 513421, ISIS 513454, ISIS 513455, ISIS 513456, ISIS 513457, ISIS 513462, ISIS 513463, ISIS 513487, ISIS 513504, ISIS 513508, and ISIS 513642.
  • One group of male BALB/c mice was injected subcutaneously twice a week for 3 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, albumin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY).
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 407935, ISIS 416472, ISIS 416549, ISIS 422087, ISIS 422096, ISIS 490103, ISIS 490196, ISIS 490208, ISIS 513454, ISIS 513455, ISIS 513456, ISIS 513457, ISIS 513487, ISIS 513504, and ISIS 513508 were considered very tolerable in terms of liver function.
  • ISIS 422086, ISIS 490209, ISIS 513419, ISIS 513420, and ISIS 513463 were considered tolerable in terms of liver function.
  • Human Target-X primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • IC 50 half maximal inhibitory concentration of each oligonucleotide is also presented in Table 27. As illustrated in Table 27, Target-X mRNA levels were reduced in a dose-dependent manner in antisense oligonucleotide treated cells. Many of the newly designed antisense oligonucleotides provided in Table 27 achieved an IC 50 of less than 0.9 ⁇ M and, therefore, are more potent than ISIS 407939.
  • Example 14 Modified antisense oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. Also tested was ISIS 407939, a 5-10-5 MOE gapmer targeting human Target-X, which was described in an earlier publication ( WO 2009/061851 ). ISIS 472998, ISIS 492878, and ISIS 493201 and 493182, 2-10-2 cEt gapmers, described in the Examples above were also included in the screen.
  • the newly designed modified antisense oligonucleotides are 16 nucleotides in length and their motifs are described in Table 28.
  • the chemistry column of Table 28 presents the sugar motif of each oligonucleotide, wherein “e” indicates a 2'-O-methoxyethyl (2'-MOE) nucleoside, "k” indicates a 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) and "d” indicates a 2'-deoxyribonucleoside.
  • Each gapmer listed in Table 28 is targeted to the human Target-X genomic sequence.
  • Example 15 Tolerability of modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X in BALB/c mice
  • mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • the newly designed modified antisense oligonucleotides were also added to this screen.
  • the newly designed chimeric antisense oligonucleotides are 16 nucleotides in length and their motifs are described in Table 29.
  • the chemistry column of Table 29 presents the sugar motif of each oligonucleotide, wherein "e” indicates a 2'-O-methoxyethyl (2'-MOE) nucleoside, "k” indicates a 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) and "d” indicates a 2'-deoxyribonucleoside.
  • Table 29 Modified chimeric antisense oligonucleotides targeted to Target-X ISIS No Chemistry SEQ CODE 516044 eee-d(10)-kkk 21 516045 eee-d(10)-kkk 22 516058 eee-d(10)-kkk 26 516059 eee-d(10)-kkk 27 516060 eee-d(10)-kkk 28 516061 eee-d(10)-kkk 29 516062 ee-d(10)-kkk 30 516046 eee-d(10)-kkk 37 516063 ee-d(10)-kkk 38 516064 eee-d(10)-kkk 89 516065 eee-d(10)-kkk 262 516066 eee-d(10)-kkk 263 516047 eee-d(10)-kkk 41 516048 eee-
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, albumin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY).
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 457851, ISIS 515635, ISIS 515637, ISIS 515638, ISIS 515643, ISIS 515647, ISIS 515649, ISIS 515650, ISIS 515652, ISIS 515654, ISIS 515656, ISIS 516056, and ISIS 516057 were considered tolerable in terms of liver function.
  • Example 16 Efficacy of modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X in transgenic mice
  • mice were developed at Taconic farms harboring a Target-X genomic DNA fragment. The mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for efficacy.
  • mice Groups of 3-4 male and female transgenic mice were injected subcutaneously twice a week for 3 weeks with 20 mg/kg/week of ISIS 457851, ISIS 515636, ISIS 515639, ISIS 515653, ISIS 516053, ISIS 516065, and ISIS 516066.
  • One group of mice was injected subcutaneously twice a week for 3 weeks with control oligonucleotide, ISIS 141923 (CCTTCCCTGAAGGTTCCTCC, 5-10-5 MOE gapmer with no known murine target, SEQ ID NO: 9).
  • ISIS 141923 control oligonucleotide
  • mice was injected subcutaneously twice a week for 3 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • Target-X ELISA kit purchased from Hyphen Bio-Med. Results are presented as percent inhibition of Target-X, relative to control. As shown in Table 31, several antisense oligonucleotides achieved reduction of human Target-X protein expression over the PBS control. Table 31 Percent inhibition of Target-X protein levels in transgenic mice ISIS No % inhibition 141923 0 457851 64 515636 68 515639 46 515653 0 516053 19 516065 0 516066 7
  • Example 17 Efficacy of modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X in transgenic mice
  • mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for efficacy.
  • mice Groups of 2-4 male and female transgenic mice were injected subcutaneously twice a week for 3 weeks with 10 mg/kg/week of ISIS 407935, ISIS 416472, ISIS 416549, ISIS 422087, ISIS 422096, ISIS 473137, ISIS 473244, ISIS 473326, ISIS 473327, ISIS 473359, ISIS 473392, ISIS 473393, ISIS 473547, ISIS 473567, ISIS 473589, ISIS 473630, ISIS 484559, ISIS 484713, ISIS 490103, ISIS 490196, ISIS 490208, ISIS 513419, ISIS 513454, ISIS 513455, ISIS 513456, ISIS 513457, ISIS 513487, ISIS 513508, ISIS 515640, ISIS 515641, ISIS 515642, ISIS 515648, ISIS 515655, ISIS 515657, ISIS 516045, ISIS 516046, ISIS 516047,
  • Antisense oligonucleotides exhibiting in vitro inhibition of Target-X mRNA were selected and tested at various doses in Hep3B cells. Also tested was ISIS 407939, a 5-10-5 MOE gapmer targeting human Target-X, which was described in an earlier publication ( WO 2009/061851 ).
  • Human Target-X primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • IC 50 half maximal inhibitory concentration of each oligonucleotide is also presented in Table 33. As illustrated in Table 33, Target-X mRNA levels were reduced in a dose-dependent manner in antisense oligonucleotide treated cells. Many of the newly designed antisense oligonucleotides provided in Table 33 achieved an IC 50 of less than 2.0 ⁇ M and, therefore, are more potent than ISIS 407939.
  • Example 19 Modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro.
  • ISIS 472998, ISIS 515652, ISIS 515653, ISIS 515654, ISIS 515655, ISIS 515656, and ISIS 515657, described in the Examples above were also included in the screen.
  • the newly designed chimeric antisense oligonucleotides are 16 or 17 nucleotides in length and their motifs are described in Table 34.
  • the chemistry column of Table 34 presents the sugar motif of each oligonucleotide, wherein “e” indicates a 2'-O-methoxyethyl (2'-MOE) nucleoside, "k” indicates a 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) and "d” indicates a 2'-deoxyribonucleoside.
  • Each gapmer listed in Table 34 is targeted to the human Target-X genomic sequence.
  • Human primer probe set RTS2927 (described hereinabove in Example 1) was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • Example 20 Design of modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) or 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications
  • additional antisense oligonucleotides were designed targeting a Target-X nucleic acid targeting start positions 1147, 1154 or 12842 of Target-X.
  • the newly designed chimeric antisense oligonucleotides are 16 or 17 nucleotides in length and their motifs are described in Table 35.
  • the chemistry column of Table 35 presents the sugar motif of each oligonucleotide, wherein "e” indicates a 2'-O-methoxyethyl (2'-MOE) nucleoside, "k” indicates a 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) and "d” indicates a 2'-deoxyribonucleoside.
  • Table 35 Chimeric antisense oligonucleotides targeted to Target-X ISIS No Chemistry SEQ CODE 529544 eek-d(10)-kke 21 529569 kee-d(10)-kke 21 529594 edk-d(10)-kke 21 529619 kde-d(10)-kke 21 529644 kddk-d(9)-kke 21 529669 kdde-d(9)-kke 21 529694 eddk-d(9)-kke 21 529929 eee-d(9)-kke 21 529809 k-d(10)-kekee 488 529828 k-d(10)-kdkee 488 529730 ke-d(10)-kekeke 489 529750 ek-d(10)-keke 489 529770 ke-d(10)-kdke 489 529790 ek-d(10)-kdke 4
  • Example 21 Modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X
  • Additional antisense oligonucleotides were designed targeting a Target-X nucleic acid and were tested for their effects on Target-X mRNA in vitro. ISIS 472998 and ISIS 515554, described in the Examples above were also included in the screen.
  • the newly designed chimeric antisense oligonucleotides are 16 nucleotides in length and their motifs are described in Table 36.
  • the chemistry column of Table 36 presents the sugar motif of each oligonucleotide, wherein “e” indicates a 2'-O-methoxyethyl (2'-MOE) nucleoside, "k” indicates a 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) and "d” indicates a 2'-deoxyribonucleoside.
  • Each gapmer listed in Table 36 is targeted to the human Target-X genomic sequence.
  • Example 22 Modified antisense oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X targeting intronic repeats
  • the newly designed chimeric antisense oligonucleotides and their motifs are described in Table 37.
  • Nucleosides followed by “d” indicate 2'-deoxyribonucleosides.
  • Nucleosides followed by “k” indicate 6'-(S)-CH 3 bicyclic nucleosides (e.g cEt).
  • Nucleosides followed by "e” indicate 2'-O-methoxyethyl (2'-MOE) nucleosides.
  • “N” indicates modified or naturally occurring nucleobases (A, T, C, G, U, or 5-methyl C).
  • Target-X Each gapmer listed in Table 37 is targeted to the intronic region of human Target-X genomic sequence, designated herein as Target-X.
  • Example 23 High dose tolerability of modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X in BALB/c mice
  • mice were treated at a high dose with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • the newly designed antisense oligonucleotides were created with the same sequences as the antisense oligonucleotides from the study described above and were also added to this screen targeting intronic repeat regions of Target-X.
  • the newly designed modified antisense oligonucleotides and their motifs are described in Table 38.
  • Nucleosides followed by “d” indicate 2'-deoxyribonucleosides.
  • Nucleosides followed by “k” indicate 6'-(S)-CH3 bicyclic nucleoside (e.g cEt) nucleosides.
  • Nucleosides followed by "e” indicate 2'-O-methoxyethyl (2'-MOE) nucleosides.
  • “N” indicates modified or naturally occurring nucleobases (A, T, C, G, U, or 5-methyl C).
  • Target-X Each gapmer listed in Table 38 is targeted to the intronic region of human Target-X genomic sequence, designated herein as Target-X.
  • Start site indicates the 5'-most nucleoside to which the gapmer is targeted in the human gene sequence.
  • Stop site indicates the 3'-most nucleoside to which the gapmer is targeted human gene sequence.
  • mice Male BALB/c mice were injected subcutaneously with a single dose of 200 mg/kg of ISIS 422142, ISIS 457851, ISIS 473294, ISIS 473295, ISIS 473327, ISIS 484714, ISIS 515334, ISIS 515338, ISIS 515354, ISIS 515366, ISIS 515380, ISIS 515381, ISIS 515382, ISIS 515384, ISIS 515386, ISIS 515387, ISIS 515388, ISIS 515406, ISIS 515407, ISIS 515408, ISIS 515422, ISIS 515423, ISIS 515424, ISIS 515532, ISIS 515533, ISIS 515534, ISIS 515538, ISIS 515539, ISIS 515558, ISIS 515656, ISIS 515575, ISIS 515926, ISIS 515944, ISIS 515945, ISIS 515948, ISIS 515949, ISIS 515951, ISIS 515952, ISSI
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, albumin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY).
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 529173, ISIS 529854, ISIS 529614, ISIS 515386, ISIS 515388, ISIS 515949, ISIS 544817, and ISIS 545479 were considered tolerable in terms of liver function.
  • Example 24 Tolerability of antisense oligonucleotides targeting human Target-X in Sprague-Dawley rats
  • Sprague-Dawley rats are a multipurpose model used for safety and efficacy evaluations.
  • the rats were treated with ISIS antisense oligonucleotides from the studies described in the Examples above and evaluated for changes in the levels of various plasma chemistry markers.
  • ALT alanine transaminase
  • AST aspartate transaminase
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 473286, ISIS 473547, ISSI 473589, ISIS 473630, ISIS 484559, ISIS 515636, ISIS 515640, ISIS 515655, ISIS 516046, and ISIS 516051 were considered very tolerable in terms of liver function.
  • ISIS 473567, ISIS 515641, ISIS 515657, ISIS 516048, and ISIS 516051 were considered tolerable in terms of liver function.
  • Example 25 Tolerability of chimeric antisense oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) modifications targeting human Target-X in Sprague-Dawley rats
  • Sprague-Dawley rats were treated with ISIS antisense oligonucleotides from the studies described in the Examples above and evaluated for changes in the levels of various plasma chemistry markers.
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on hepatic function, plasma levels of transaminases were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY). Plasma levels of Bilirubin and BUN were also measured using the same clinical chemistry analyzer.
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 416507, ISIS 490208, ISIS 490368, ISIS 490396, ISIS 490803, ISIS 491122, ISIS 513446, ISIS 513454, ISIS 513455, ISIS 513456, ISIS 513504, and ISIS 513508 were considered very tolerable in terms of liver function.
  • ISIS 407936, ISIS 416508, ISIS 490279, and ISIS 513507 were considered tolerable in terms of liver function.
  • Example 26 Tolerability of chimeric antisense oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) modifications targeting human Target-X in CD-1 mice
  • CD-1 mice are a multipurpose mice model, frequently utilized for safety and efficacy testing.
  • the mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, albumin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY).
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 473295, ISIS 473714, ISIS 515558, ISIS 515926, 515951, ISIS 515952, ISIS 529126, ISIS 529166, 529564, ISIS 529582, ISIS 529614, ISIS 529725, ISIS 529765, ISIS 529799, ISIS 529823, and ISIS 534594 were considered very tolerable in terms of liver function.
  • ISIS 515424, ISIS 515534, ISIS 515926, ISIS 529785, and ISIS 534664 were considered tolerable in terms of liver function.
  • Example 27 Tolerability of chimeric antisense oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) modifications targeting human Target-X in CD-1 mice
  • CD-1 mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • Groups of 3 male CD-1 mice each were injected subcutaneously twice a week for 6 weeks with 100 mg/kg of ISIS 490208, ISIS 490279, ISIS 490323, ISIS 490368, ISIS 490396, ISIS 490803, ISIS 491122, ISIS 513419, ISIS 513446, ISIS 513454, ISIS 513455, ISIS 513456, ISIS 513504, ISIS 513507, and ISIS 513508.
  • Groups of 3 male CD-1 mice each were injected subcutaneously twice a week for 6 weeks with 100 mg/kg of ISIS 407936, ISIS 416507, and ISIS 416508, which are gapmers described in a previous publication.
  • One group of male CD-1 mice was injected subcutaneously twice a week for 6 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY).
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 407936, ISIS 416507, ISIS 490279, ISIS 490368, ISIS 490396, ISIS 490803, ISIS 491122, ISIS 513446, ISIS 513454, ISIS 513456, and ISIS 513504 were considered very tolerable in terms of liver function.
  • ISIS 490208, ISIS 513455, ISIS 513507, and ISIS 513508 were considered tolerable in terms of liver function.
  • Example 28 Efficacy of modified oligonucleotides comprising 2'-O-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X in transgenic mice
  • mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for efficacy.
  • mice Groups of 2-3 male and female transgenic mice were injected subcutaneously twice a week for 3 weeks with 5 mg/kg/week of ISIS 473244, ISIS 473295, ISIS 484714, ISIS 515926, ISIS 515951, ISIS 515952, ISIS 516062, ISIS 529126, ISIS 529553, ISIS 529745, ISIS 529799, ISIS 534664, ISIS 534826, ISIS 540168, ISIS 540175, ISIS 544826, ISIS 544827, ISIS 544828, and ISIS 544829.
  • One group of mice was injected subcutaneously twice a week for 3 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • Plasma protein levels of Target-X were estimated using a Target-X ELISA kit (purchased from Hyphen Bio-Med). Results are presented as percent inhibition of Target-X, relative to control. As shown in Table 39, several antisense oligonucleotides achieved reduction of human Target-X over the PBS control. 'n.d.' indicates that the value for that particular oligonucleotide was not measured. Table 39 Percent inhibition of Target-X plasma protein levels in transgenic mice ISIS No % inhibition 473244 2 473295 13 484714 19 515926 11 515951 13 515952 0 516062 62 529126 0 529553 0 529745 22 529799 26 534664 32 534826 n.d. 540168 94 540175 98 544813 0 544826 23 544827 60 544828 33 544829 53
  • Example 29 Efficacy of modified oligonucleotides comprising 2'-methoxyethyl (2'-MOE) and 6'-(S)-CH 3 bicyclic nucleoside (e.g cEt) modifications targeting human Target-X in transgenic mice
  • mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for efficacy.
  • mice Groups of 2-3 male and female transgenic mice were injected subcutaneously twice a week for 3 weeks with 1 mg/kg/week of ISIS 407936, ISIS 490197, ISIS 490275, ISIS 490278, ISIS 490279, ISIS 490323, ISIS 490368, ISIS 490396, ISIS 490803, ISIS 491122, ISIS 513446, ISIS 513447, ISIS 513504, ISIS 516062, ISIS 529166, ISIS 529173, ISIS 529360, ISIS 529725, ISIS 534557, ISIS 534594, ISIS 534664, ISIS 534688, ISIS 534689, ISIS 534915, ISIS 534916, ISIS 534917, and ISIS 534980.
  • One group of mice was injected subcutaneously twice a week for 3 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • Plasma protein levels of Target-X were estimated using a Target-X ELISA kit (purchased from Hyphen Bio-Med). Results are presented as percent inhibition of Target-X, relative to control. As shown in Table 40, several antisense oligonucleotides achieved reduction of human Target-X over the PBS control.
  • Example 30 Tolerability of antisense oligonucleotides targeting human Target-X in Sprague-Dawley rats
  • Sprague-Dawley rats were treated with ISIS antisense oligonucleotides from the studies described in the Examples above and evaluated for changes in the levels of various plasma chemistry markers.
  • ALT alanine transaminase
  • AST aspartate transaminase
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused increase in the levels within three times the upper limit of normal levels of transaminases were deemed very tolerable.
  • ISIS oligonucleotides that caused increase in the levels of transaminases between three times and seven times the upper limit of normal levels were deemed tolerable. Based on these criteria, ISIS 515380, ISIS 515387, ISIS 529175, ISIS 529176, ISIS 529804, and ISIS 537064 were considered very tolerable in terms of liver function. Based on these criteria, ISIS 515381 was considered tolerable in terms of liver function.
  • Example 31 Efficacy of antisense oligonucleotides targeting human Target-X in transgenic mice
  • mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for efficacy.
  • mice Two groups of 3 male and female transgenic mice were injected subcutaneously twice a week for 2 weeks with 0.5 mg/kg/week or 1.5 mg/kg/week of ISIS 407935 and ISIS 513455. Another group of mice was subcutaneously twice a week for 2 weeks with 0.6 mg/kg/week or 2.0 mg/kg/week of ISIS 473286.
  • mice were subcutaneously twice a week for 2 weeks with 0.1 mg/kg/week or 0.3 mg/kg/week of ISIS 473589, ISIS 515380, ISIS 515423, ISIS 529804, ISIS 534676, ISIS 534796, ISIS 540162, ISIS 540164, ISIS 540175, ISIS 540179, ISIS 540181, ISIS 540182, ISIS 540186, ISIS 540191, ISIS 540193, ISIS 544827, or ISIS 545474.
  • Another 3 groups of mice were injected subcutaneously twice a week for 2 weeks with 0.3 mg/kg/week of ISIS 516062, ISIS 534528 or ISIS 534693.
  • One group of mice was injected subcutaneously twice a week for 2 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • Plasma protein levels of Target-X were estimated using a Target-X ELISA kit (purchased from Hyphen Bio-Med). Results are presented as percent inhibition of Target-X, relative to control. As shown in Table 41, several antisense oligonucleotides achieved reduction of human Target-X over the PBS control.
  • Example 32 Tolerability of antisense oligonucleotides targeting human Target-X in Sprague-Dawley rats
  • ALT alanine transaminase
  • AST aspartate transaminase
  • ISIS oligonucleotides that did not cause any increase in the levels of transaminases, or which caused an increase within three times the upper limit of normal (ULN) were deemed very tolerable.
  • ISIS oligonucleotides that caused an increase in the levels of transaminases between three times and seven times the ULN were deemed tolerable.
  • ISIS 540164, ISIS 540172, and ISIS 540175 were considered very tolerable in terms of liver function.
  • ISIS 534676, ISIS 534796, ISIS 534797, ISIS 540162, and ISIS 540179 were considered tolerable in terms of liver function.
  • Antisense oligonucleotides selected from the studies described above were tested at various doses in Hep3B cells.
  • Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.05 ⁇ M, 0.15 ⁇ M, 0.44 ⁇ M, 1.33 ⁇ M, and 4.00 ⁇ M concentrations of antisense oligonucleotide, as specified in Table 42.
  • RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR.
  • Human Target-X primer probe set RTS2927 was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells.
  • IC 50 half maximal inhibitory concentration
  • Example 34 Tolerability of antisense oligonucleotides targeting human Target-X in CD-1 mice
  • CD-1 mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • mice Two groups of 4 male 6-8 week old CD-1 mice each were injected subcutaneously twice a week for 6 weeks with 50 mg/kg of ISIS 407935 and ISIS 490279. Another seven groups of 4 male 6-8 week old CD-1 mice each were injected subcutaneously twice a week for 6 weeks with 25 mg/kg of ISIS 473589, ISIS 529804, ISIS 534796, ISIS 540162, ISIS 540175, ISIS 540182, and ISIS 540191. One group of male CD-1 mice was injected subcutaneously twice a week for 6 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, albumin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY). The results are presented in Table 43. Treatment with the newly designed antisense oligonucleotides were more tolerable compared to treatment with ISIS 407935 (disclosed in an earlier publication), which caused elevation of ALT levels greater than seven times the upper limit of normal (ULN).
  • ISIS 407935 Disclosed in an earlier publication
  • Body weights, as well as liver, heart, lungs, spleen and kidney weights were measured at the end of the study, and are presented in Table 44.
  • ISIS oligonucleotides did not cause any changes in organ weights outside the expected range and were therefore deemed tolerable in terms of organ weights.
  • Example 35 Tolerability of antisense oligonucleotides targeting human Target-X in Sprague-Dawley rats
  • Sprague-Dawley rats were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various plasma chemistry markers.
  • ISIS oligonucleotides To evaluate the effect of ISIS oligonucleotides on liver and kidney function, plasma levels of transaminases, bilirubin, albumin, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, NY). The results are presented in Table 45. Treatment with the all antisense oligonucleotides was tolerable in terms of plasma chemistry markers in this model.
  • Body weights, as well as liver, heart, lungs, spleen and kidney weights were measured at the end of the study, and are presented in Table 46. Treatment with all the antisense oligonucleotides was tolerable in terms of body and organ weights in this model.
  • Example 36 Dose-dependent antisense inhibition of human Target-X in cynomolgos monkey primary hepatocytes
  • Antisense oligonucleotides selected from the studies described above were tested at various doses in cynomolgous monkey primary hepatocytes.
  • Cells were plated at a density of 35,000 cells per well and transfected using electroporation with 0.009 ⁇ M, 0.03 ⁇ M, 0.08 ⁇ M, 0.25 ⁇ M, 0.74 ⁇ M, 2.22 ⁇ M, 6.67 ⁇ M, and 20.00 ⁇ M concentrations of antisense oligonucleotide, as specified in Table 47.
  • RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR.
  • Target-X primer probe set RTS2927 was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells. As illustrated in Table 47, Target-X mRNA levels were reduced in a dose-dependent manner with some of the antisense oligonucleotides that are cross-reactive with the rhesus monkey genomic sequence.
  • Antisense oligonucleotides from the study described above were also tested at various doses in Hep3B cells.
  • Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.009 ⁇ M, 0.03 ⁇ M, 0.08 ⁇ M, 0.25 ⁇ M, 0.74 ⁇ M, 2.22 ⁇ M, 6.67 ⁇ M, and 20.00 ⁇ M concentrations of antisense oligonucleotide, as specified in Table 48.
  • RNA was isolated from the cells and Target-X mRNA levels were measured by quantitative real-time PCR.
  • Target-X primer probe set RTS2927 was used to measure mRNA levels.
  • Target-X mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN ® . Results are presented as percent inhibition of Target-X, relative to untreated control cells. As illustrated in Table 48, Target-X mRNA levels were reduced in a dose-dependent manner with several of the antisense oligonucleotides.
  • Example 38 Efficacy of antisense oligonucleotides targeting human Target-X in transgenic mice
  • mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for efficacy.
  • mice Eight groups of 3 transgenic mice each were injected subcutaneously twice a week for 3 weeks with 20 mg/kg/week, 10 mg/kg/week, 5 mg/kg/week, or 2.5 mg/kg/week of ISIS 407935 or ISIS 490279. Another 24 groups of 3 transgenic mice each were subcutaneously twice a week for 3 weeks with 5 mg/kg/week, 2.5 mg/kg/week, 1.25 mg/kg/week, or 0.625 mg/kg/week of ISIS 473589, ISIS 529804, ISIS 534796, ISIS 540162, ISIS 540175, or ISIS 540191. One group of mice was injected subcutaneously twice a week for 3 weeks with PBS. Mice were euthanized 48 hours after the last dose, and organs and plasma were harvested for further analysis.
  • Plasma protein levels of Target-X were estimated using a Target-X ELISA kit (purchased from Hyphen Bio-Med). As shown in Table 50, several antisense oligonucleotides achieved reduction of human Target-X over the PBS control. Results are presented as percent inhibition of Target-X, relative to control.
  • Cynomolgus monkeys were treated with ISIS antisense oligonucleotides selected from studies described above, including ISIS 407935, ISIS 490279, ISIS 473589, ISIS 529804, ISIS 534796, ISIS 540162, ISIS 540175, ISIS 540182, and ISIS 540191. Antisense oligonucleotide efficacy was evaluated. ISIS 407935, from the earlier publication, was included in the study for comparison.
  • the monkeys Prior to the study, the monkeys were kept in quarantine for at least a 30-day period, during which the animals were observed daily for general health. Standard panels of serum chemistry and hematology, examination of fecal samples for ova and parasites, and a tuberculosis test were conducted immediately after the animals' arrival to the quarantine area. The monkeys were 2-4 years old at the start of treatment and weighed between 2 and 4 kg. Ten groups of four randomly assigned male cynomolgus monkeys each were injected subcutaneously with ISIS oligonucleotide or PBS using a stainless steel dosing needle and syringe of appropriate size into one of 4 sites on the back of the monkeys; each site used in clock-wise rotation per dose administered.
  • Plasma Target-X levels were measured prior to dosing, and on day 3, day 5, day 7, day 16, day 30, day 44, day 65, and day 86 of treatment.
  • Target-X activity was measured using Target-X deficiuent plasma.
  • Approximately 1.5 mL of blood was collected from all available study animals into tubes containing 3.2% sodium citrate. The samples were placed on ice immediately after collection. Collected blood samples were processed to platelet poor plasma and the tubes were centrifuged at 3,000 rpm for 10 min at 4°C to obtain plasma.
  • Target-X protein levels were measured by a Target-X elisa kit (purchased from Hyphen BioMed). The results are presented in Table 53.
  • Table 53 Plasma Target-X protein levels (% reduction compared to the baseline) in the cynomolgus monkey plasma ISIS No Day 3 Day 5 Day 7 Day 16 Day 30 Day 44 Day 65 Day 86 407935 21 62 69 82 84 85 84 90 490279 0 29 35 30 38 45 51 58 473589 12 67 85 97 98 98 98 529804 19 65 76 87 88 89 90 90 534796 1 46 54 64 64 67 66 70 540162 0 24 26 37 45 49 49 50 540175 0 28 36 38 47 52 55 55 540182 0 17 8 0 0 0 5 0 540191 0 12 4 0 0 4 9 10
  • SNPs Single nucleotide polymorphisms in the huntingtin ( HTT ) gene sequence
  • SNP positions (identified by Hayden et al, WO/2009/135322 ) associated with the HTT gene were mapped to the HTT genomic sequence, designated herein as SEQ ID NO: 1 (NT_006081.18 truncated from nucleotides 1566000 to 1768000).
  • Table 56 provides SNP positions associated with the HTT gene.
  • the 'Reference SNP ID number' or 'RS number' is the number designated to each SNP from the Entrez SNP database at NCBI, incorporated herein by reference.
  • 'SNP position' refers to the nucleotide position of the SNP on SEQ ID NO: 1.
  • 'Polymorphism' indicates the nucleotide variants at that SNP position.
  • 'Major allele' indicates the nucleotide associated with the major allele, or the nucleotide present in a statistically significant proportion of individuals in the human population.
  • 'Minor allele' indicates the nucleotide associated with the minor allele, or the nucleotide present in a relatively small proportion of individuals in the human population.
  • SNPs Single Nuclear Polymorphisms
  • SEQ ID N O 1 RS No. SNP position Polymorphism Major allele Minor allele rs2857936 1963 C/T C T rs12506200 3707 A/G G A rs762855 14449 A/G G A rs3856973 19826 G/A G A rs2285086 28912 G/A A G rs7659144 37974 C/G C G rs16843804 44043 C/T C T rs2024115 44221 G/A A G rs10015979 49095 A/G A G rs7691627 51063 A/G G A rs2798235 54485 G/A G A rs4690072 62160 G/T T G rs6446723 66466 C/T T C rs363081 73280 G/A G A rs363080 73564 T/C C T r
  • HTT Huntingtin
  • SNP Single Nucleotide Polymorphism
  • modified oligonucleotides were designed based on the parent gapmer, ISIS 460209 wherein the central gap region contains nine 2'-deoxyribonucleosides. These modified oligonucleotides were designed by introducing various chemical modifications in the central gap region and were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting rs7685686 while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the modified oligonucleotides were evaluated and compared to the parent gapmer, ISIS 460209.
  • the modified oligonucleotides were created with a 3-9-3 motif and are described in Table 57.
  • All cytosine nucleobases thoughout each gapmer are 5-methyl cytosines.
  • Nucleosides without a subscript are ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e”, "k”, “y”, or "z” are sugar modified nucleosides.
  • a subscript "e” indicates a 2'-O-methoxyethyl (MOE) modified nucleoside
  • a subscript "k” indicates a 6'- (S) -CH 3 bicyclic nucleoside (e.g. cEt)
  • a subscript "y” indicates an ⁇ -L-LNA bicyclic nucleoside
  • a subscript "z” indicates a F-HNA modified nucleoside.
  • p U indicates a 5-propyne uridine nucleoside and x T indicates a 2-thio-thymidine nucleoside.
  • the number in parentheses indicates the position on the modified oligonucleotide opposite to the SNP position, as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used (from Coriell Institute).
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented below.
  • the half maximal inhibitory concentration (IC 50 ) of each oligonucleotide is presented in Table 58 and was calculated by plotting the concentrations of oligonucleotides used versus the percent inhibition of HTT mRNA expression achieved at each concentration, and noting the concentration of oligonucleotide at which 50% inhibition of HTT mRNA expression was achieved compared to the control.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • the parent gapmer, ISIS 460209 is marked with an asterisk (*) in the table and was included in the study as a benchmark oligonucleotide against which the activity and selectivity of the modified oligonucleotides targeting nucleotides overlapping the SNP position could be compared.
  • modified oligonucleotides having chemical modifications in the central gap region at the SNP position exhibited similar activity with an increase in selectivity comparing to the parent gapmer, wherein the central gap region contains full deoxyribonucleosides.
  • Table 57 Modified oligonucleotides targeting HTT rs7685686 ISIS NO Sequence (5' to 3') Gap chemistry Wing chemistry SEQ ID NO.
  • HTT Huntingtin
  • SNP Single Nucleotide Polymorphism
  • Additional modified oligonucleotides were designed in a similar manner as the antisense oligonucleotides described in Table 57. Various chemical modifications were introduced in the central gap region at the SNP position in an effort to improve selectivity while maintaining activity in reducing mutant HTT mRNA levels.
  • the modified oligonucleotides were created with a 3-9-3 motif and are described in Table 59.
  • All cytosine nucleobases thoughout each gapmer are 5-methyl cytosines.
  • Nucleosides without a subscript are ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "a”, “e”, “f”, “h”, “k”, “I”, “R”, “w” are sugar modified nucleosides.
  • a subscript "a” indicates a 2'-(ara)-F modified nucleoside
  • a subscript "e” indicates a 2'-O-methoxyethyl (MOE) modified nucleoside
  • a subscript "f” indicates a 2'-F modified nucleoside
  • a subscript "h” indicates a HNA modified nucleoside
  • a subscript "k” indicates a 6'- (S) -CH 3 bicyclic nucleoside (e.g.
  • n T indicates an N3-ethylcyano thymidine nucleoside
  • b N indicates an abasic nucleoside (e.g. 2'-deoxyribonucleoside comprising a H in place of a nucleobase).
  • Underlined nucleoside or the number in parentheses indicates the position on the modified oligonucleotide opposite to the SNP position, as counted from the 5'-terminus.
  • the modified oligonucleotides were evaluated in thermal stability (T m ) assay.
  • T m thermal stability
  • the T m 's were measured using the method described herein.
  • a Cary 100 Bio spectrophotometer with the Cary Win UV Thermal program was used to measure absorbance vs. temperature.
  • oligonucleotides were prepared at a concentration of 8 ⁇ M in a buffer of 100 mM Na+, 10 mM phosphate, 0.1 mM EDTA, pH 7. Concentration of oligonucleotides were determined at 85 °C. The oligonucleotide concentration was 4 ⁇ M with mixing of equal volumes of test oligonucleotide and mutant or wild-type RNA strand.
  • Oligonucleotides were hybridized with the mutant or wild-type RNA strand by heating duplex to 90 °C for 5 min and allowed to cool at room temperature. Using the spectrophotometer, T m measurements were taken by heating duplex solution at a rate of 0.5 C/min in cuvette starting @ 15 °C and heating to 85 °C. T m values were determined using Vant Hoff calculations (A 260 vs temperature curve) using non self-complementary sequences where the minimum absorbance which relates to the duplex and the maximum absorbance which relates to the non-duplex single strand are manually integrated into the program.
  • T m for the modified oligonucleotides when duplexed to mutant or wild-type RNA complement.
  • the T m of the modified oligonucleotides duplexed with mutant RNA complement is denoted as "T m (°C) mut”.
  • the T m of the modified oligonucleotides duplexed with wild-type RNA complement is denoted as "T m (°C) wt".
  • the modified oligonucleotides were also tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with a single dose at 2 ⁇ M concentration of the modified oligonucleotide. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele.. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. The results in Table 60 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC". Selectivity as was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of mutant HTT mRNA levels.
  • the parent gapmer, ISIS 460209 is marked with an asterisk (*) in the table and was included in the study as a benchmark oligonucleotide against which the selectivity of the modified oligonucleotides targeting nucleotides overlapping the SNP position could be compared.
  • Modified oligonucleotides comprising modified nucleobase, N3-ethylcyano (ISIS 539564) or abasic nucleobase (ISIS 543525) showed little to no improvement in selectivity.
  • Table 59 Modified oligonucleotides comprising chemical modifications in the central gap region ISIS NO Sequence (5' to 3') Gap chemistry Wing chemistry SEQ ID NO.
  • Chimeric oligonucleotides were designed based on the parent gapmer, ISIS 460209. These gapmers comprise self-complementary regions flanking the central gap region, wherein the central gap region contains nine deoxyribonucleosides and the self-complementary regions are complementary to one another.
  • the underlined nucleosides indicate the portion of the 5'-end that is self-complement to the portion of the 3'-end.
  • the gapmers and their motifs are described in Table 61.
  • MOE 2'-O-methoxyethyl
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with a single dose at 2 ⁇ M concentration of the modified oligonucleotide. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. The results in Table 62 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC". Selectivity was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of the mutant HTT mRNA levels.
  • the parent gapmer, ISIS 460209 is marked with an asterisk (*) in the table and was included in the study as a benchmark oligonucleotide against which the selectivity of the modified oligonucleotides targeting nucleotides overlapping the SNP position could be compared.
  • Gapmers are designed based on the most selective gapmers from studies described in Tables 61 and 62 (ISIS 550912 and 550913). These gapmers are created such that they cannot form self-structure in the effort to evaluate if the increased activity simply is due to higher binding affinity. Gapmers are designed by deleting two or three nucleotides at the 3'- terminus and are created with 6-9-3 or 5-9-3 motif.
  • the chimeric oligonucleotides and their motifs are described in Table 63.
  • MOE 2'-O-methoxyethyl
  • the gapmers, ISIS 550912 and ISIS 550913, from which the newly designed gapmers are derived from, are marked with an asterisk (*) in the table.
  • Table 63 Non-self-complementary chimeric oligonucleotides targeting HTT SNP ISIS NO Sequence (5' to 3') Motif Wing chemistry SEQ ID NO.
  • a series of chimeric antisense oligonucleotides were designed based on the parent gapmer, ISIS 460209, wherein the central gap region contains nine 2'-deoxyribonucleosides. These gapmers were designed by introducing modified nucleosides at both 5' and 3' termini. Gapmers were also created with a single mismatch shifted slightly upstream and downstream (i.e. "microwalk") within the central gap region and with the SNP position opposite position 5 of the parent gapmer, as counted from the 5'-gap terminus.
  • microwalk slightly upstream and downstream
  • the gapmers and their motifs are described in Table 64.
  • MOE 2'-O-methoxyethyl
  • k indicates a 6'- (S) -CH 3 bicyclic
  • T m thermal stability
  • Heterozygous fibroblast GM04022 cell line was used. Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with a single dose at 2 ⁇ M concentration of the modified oligonucleotide. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed. RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. The results in Table 65 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC". Selectivity was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of mutant HTT mRNA levels.
  • the parent gapmer, ISIS 460209 is marked with an asterisk (*) in the table and was included in the study as a benchmark oligonucleotide against which the selectivity of the modified oligonucleotides targeting nucleotides overlapping the SNP position could be compared.
  • Additional chimeric antisense oligonucleotides are designed based on two gapmers selected from studies described in Tables 64 and 65 (ISIS 476333 and ISIS 460209) wherein the central gap region contains nine 2'-deoxyribonucleosides. These gapmers are designed by introducing a single mismatch, wherein the mismatch will be shifted throughout the antisense oligonucleotide (i.e. "microwalk”). Gapmers are also created with 4-9-4 or 3-9-3 motifs and with the SNP position opposite position 8 of the original gapmers, as counted from the 5'- terminus.
  • the gapmers and their motifs are described in Table 66.
  • MOE 2'-O-methoxyethyl
  • k indicates a 6'- (S) -CH 3 bicyclic nucle
  • HTT Huntingtin
  • SNP Single Nucleotide Polymorphism
  • Chimeric antisense oligonucleotides were designed based on the parent gapmer, ISIS 460209, wherein the central gap region contains nine 2'-deoxyribonucleosides. These gapmers were designed by shortening the central gap region to seven 2'-deoxyribonuclosides. Gapmers were also created with 5-7-5 motif and with the SNP position opposite position 8 or 9 of the parent gapmer, as counted from the 5'-terminus.
  • the gapmers and their motifs are described in Table 67.
  • MOE 2'-O-methoxyethyl
  • the chimeric antisense oligonucleotides were tested in vitro.
  • ISIS 141923 was included in the study as a negative control and is denoted as "neg control”.
  • a non-allele specific antisense oligonucleotide, ISIS 387916 was used as a positive control and is denoted as "pos control”.
  • ISIS 460209 was included in the study for comparison.
  • Heterozygous fibroblast GM04022 cell line was used. Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3, and 10 ⁇ M concentration of the modified oligonucleotide.
  • RT-PCR method in short A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA.
  • HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 68.
  • HTT Huntingtin
  • SNP Single Nucleotide Polymorphism
  • Additional chimeric antisense oligonucleotides were designed based on the parent gapmer, ISIS 460209, wherein the central gap region contains nine 2'-deoxyribonucleosides. These gapmers were designed with the central gap region shortened or interrupted by introducing various modifications either within the gap or by adding one or more modified nucleosides to the 3'-most 5'-region or to the 5'-most 3'-region. Gapmers were created with the SNP position opposite position 8 of the parent gapmer, as counted from the 5'- terminus.
  • the gapmers and their motifs are described in Table 69.
  • MOE 2'-O-methoxyethyl
  • chimeric antisense oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 2 ⁇ M concentration of the modified oligonucleotide. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. The results in Table 70 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC". Selectivity was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of mutant HTT mRNA levels. ISIS 460209 marked with an asterisk (*) in the table was included in the study for comparison.
  • the gapmers and their motifs are described in Table 71.
  • Subscript "p” indicates methyl phosphonate internucleoside linkage.
  • Subscript "pz” indicates (R) -methyl phosphonate internucleoside linkage.
  • Subscript "pw” indicates (S) -methyl phosphonate internucleoside linkage.
  • All cytosine nucleobases thoughout each gapmer are 5-methyl cytosines.
  • x T indicates a 2-thio thymidine nucleoside.
  • Nucleosides without a subscript are ⁇ -D-2'-deoxyribonucleosides. Nucleosides followed by a subscript “e”, “k” or “b” are sugar modified nucleosides. A subscript “e” indicates a 2'-O-methoxyethyl (MOE) modified nucleoside, a subscript “k” indicates a 6'- (S) -CH 3 bicyclic nucleoside (e.g. cEt) and a subscript "b” indicates a 5'-Me DNA modified nucleoside. Underlined nucleosides indicate the position of modification. Bold and underlined nucleosides indicate the mismatch position.
  • MOE 2'-O-methoxyethyl
  • k 6'- (S) -CH 3 bicyclic nucleoside
  • a subscript "b” indicates a 5'-Me DNA modified nucleoside.
  • Underlined nucleosides indicate the position
  • Short-gap chimeric oligonucleotides comprising modifications at the wing regions targeting Huntingtin ( HTT ) Single Nucleotide Polymorphism (SNP)
  • the gapmers and their motifs are described in Table 72.
  • MOE 2'-O-methoxyethyl
  • the number in parentheses indicates the position on the chimeric oligonucleotide opposite to the SNP position, as counted from the 5'-terminus.
  • T m thermal stability
  • Heterozygous fibroblast GM04022 cell line was used. Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with a single dose at 2 ⁇ M concentration of the modified oligonucleotide. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed. RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. The results in Table 73 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC". Selectivity was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of mutant HTT mRNA levels. ISIS 460209 marked with an asterisk (*) in the table was included in the study for comparison.
  • the gapmers and their motifs are described in Table 74.
  • P phosphorothioate
  • All cytosine nucleobases thoughout each gapmer are 5-methyl cytos
  • the SNP site indicates the position on the chimeric antisense oligonucleotide opposite to the SNP position, as counted from the 5'-gap terminus and is denoted as "SNP site”.
  • IC 50 and selectivity were calculated using the methods previously described in Example 41.
  • Table 75 chimeric oligonucleotides comprising 4-9-2 (ISIS 540082) or 2-9-4 (ISIS 540095) motif with the SNP site at position 1 or 3 showed comparable activity and 2.5 fold selectivity as compared to their counterparts.
  • Table 74 Chimeric oligonucleotides designed by microwalk ISIS NO Sequence (5' to 3') Motif SNP site wing chemistry SEQ ID NO.
  • Chimeric antisense oligonucleotides were designed based on the parent gapmer, ISIS 460209 wherein the SNP site aligns with position 8 of the parent gapmer, as counted from the 5'-terminus. These gapmers were designed by shifting the SNP site upstream or downstream (i.e. microwalk) of the original oligonucleotide.
  • the gapmers and their motifs are described in Table 76.
  • MOE 2'-O-methoxyethyl
  • chimeric oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides. After a treatment period of approximately 16 hours, cells were washed with DPBS buffer and lysed.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. The results in Table 77 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC". Selectivity was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of mutant HTT mRNA levels.
  • the parent gapmer, ISIS 460209 is marked with an asterisk (*) in the table and was included in the study as a benchmark oligonucleotide against which the selectivity of the modified oligonucleotides targeting nucleotides overlapping the SNP position could be compared.
  • a series of modified oligonucleotides were designed based on the parent gapmer, ISIS 460209, wherein the central gap region comprises nine 2'-deoxyribonucleosides. These gapmers were created with various motifs and modifications at the wings and/or the central gap region.
  • the modified oligonucleotides and their motifs are described in Table 78.
  • a subscript "e” indicates a 2'-O-methoxyethyl (MOE) modified nucleoside
  • a subscript "k” indicates a 6'- (S) -CH 3 bicyclic nucleoside (e.g. cEt)
  • a subscript "y” indicates an ⁇ -L-LNA modified nucleoside
  • a subscript "z” indicates a F-HNA modified nucleoside.
  • p U indicates a 5-propyne uridine nucleoside and x T indicates a 2-thio-thymidine nucleoside.
  • Underlined nucleosides indicate the mismatch position.
  • T m thermal stability
  • ISIS 141923 was included in the study as a negative control and is denoted as "neg control”.
  • the non-allele specific antisense oligonucleotides, ISIS 387916 was used as a positive control and is denoted as "pos control”.
  • Heterozygous fibroblast GM04022 cell line was used. Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with a single dose at 2 ⁇ M concentration of the modified oligonucleotide. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed.
  • RT-PCR method in short A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele.
  • HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. ISIS 460209 marked with an asterisk (*) in the table was included in the study for comparison. The results in Table 79 are presented as percent of HTT mRNA expression, relative to untreated control levels and is denoted as "% UTC”. Selectivity was also evaluated and measured by dividing the percent of wild-type HTT mRNA levels vs . the percent of mutant HTT mRNA levels.
  • the gapmers are described in Table 80.
  • All cytosine nucleobases thoughout each gapmer are 5-methyl cytosines.
  • Nucleosides without a subscript are ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "a”, "b", "e”, or "k” are sugar modified nucleosides.
  • a subscript "a” indicates 2'-(ara)-F modified nucleoside
  • a subscript "b” indicates a 5'-Me DNA modified nucleoside
  • a subscript "e” indicates a 2'-O-methoxyethyl (MOE) modified nucleoside
  • a subscript "k” indicates a 6'- (S) -CH 3 bicyclic nucleoside (e.g. cEt).
  • x T indicates a 2-thio-thymidine nucleoside.
  • Underline nucleoside or the number in parentheses indicates the position on the oligonucleotides opposite to the SNP position, as counted from the 5'-terminus.
  • gapmers are designed based on the gapmer selected from studies described in Tables 89 and 21 (ISIS 540107) and is marked with an asterisk (*). These gapmers are designed by introducing bicyclic modified nucleosides at the 3' or 5' terminus and are tested to evaluate if the addition of bicyclic modified nucleosides at the wing regions improves the activity and selectivity in inhibition of mutant HTT SNP.
  • Chimeric oligonucleotides comprising wing and central gap modifications targeting HTT SNP
  • Additional gapmers are designed based on the parent gapmer, ISIS 460209, wherein the central gap region comprises nine 2'-deoxyribonucleosides and is marked with an asterisk (*) in the table. These gapmers were designed by introducing modifications at the wings or the central gap region and are created with a 3-9-3 motif.
  • the gapmers are described in Table 82.
  • MOE 2'-O-methoxyethyl
  • P T indicates a 5-propyne thymidine nucleoside.
  • P C indicates a 5-propyne cytosine nucleoside.
  • Underline nucleoside or the number in parentheses indicates the position on the oligonucleotides opposite to the SNP position, as counted from the 5' -terminus.
  • Modified oligonucleotides comprising F-HNA modification at the central gap or wing region targeting HTT SNP
  • modified oligonucleotides were designed based on ISIS 460209, wherein the central gap region contains nine 2'-deoxyribonucleosides. These modified oligonucleotides were designed by incorporating one or more F-HNA(s) modification within the central gap region or on the wing regions. The F-HNA containing oligonucleotides were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting rs7685686 while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the modified oligonucleotides were evaluated and compared to ISIS 460209.
  • the modified oligonucleotides and their motifs are described in Table 83.
  • Nucleosides without a subscript are ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • Nucleosides followed by a subscript "z” indicate F-HNA modified nucleosides.
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 as count
  • the gap-interrupted antisense oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 84.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • the parent gapmer, 460209 is marked with an asterisk (*) in the table and was included in the study as a benchmark oligonucleotide against which the activity and selectivity of antisense oligonucleotides targeting nucleotides overlapping the SNP position could be compared.
  • oligonucleotides comprising F-HNA modification(s) showed improvement in selectivity while maintaining activity as compared to the parent gapmer, ISIS 460209.
  • Table 83 Gap-interrupted antisense oligonucleotides targeting HTT SNP ISIS NO. Sequence (5' to 3') Motif Gap chemistry Wing chemistry SEQ ID NO.
  • Modified oligonucleotides comprising cEt modification(s) at the central gap region targeting HTT SNP
  • modified oligonucleotides were designed in the same manner as described in Example 57. These modified oligonucleotides were designed by replacing F-HNA(s) with cEt modification(s) in the central gap region while maintaining the wing configuration. The modified oligonucleotides were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting rs7685686 while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the modified oligonucleotides were evaluated and compared to ISIS 460209.
  • the modified oligonucleotides and their motifs are described in Table 85.
  • Nucleosides without a subscript are ( ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 as counted from the 5'-terminus.
  • the gap-interrupted antisense oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented below.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • the modified oligonucleotides and their motifs are described in Table 87.
  • Nucleosides without a subscript are ( ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • Nucleosides followed by a subscript "z” indicate F-HNA modified nucleosides.
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 as count
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 88.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • Short-gap chimeric oligonucleotides targeting Huntingtin HCT
  • Single Nucleotide Polymorphism SNP
  • Additional chimeric antisense oligonucleotides were designed based on ISIS 460209 and ISIS 540094 wherein the central gap region contains nine 2'-deoxynucleosides. These gapmers were designed with the central gap region shortened by introducing cEt modifications to the wing regions, or interrupted by introducing cEt modifications at the 3'-end of the the central gap region. The modified oligonucleotides were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting HTT SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the modified oligonucleotides were evaluated and compared to ISIS 460209 and 540094.
  • the gapmers and their motifs are described in Table 89.
  • Nucleosides without a subscript are ( ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 4 or 8 as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 90.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • the newly designed antisense oligonucleotides (ISIS 575003) showed improvement in selectivity while maintaining potency as compared to ISIS 460209.
  • Table 89 Short-gap antisense oligonucleotides targeting HTT SNP ISIS NO. Sequence (5' to 3') Motif Gap chemistry Wing chemistry SEQ ID NO.
  • Short-gap chimeric oligonucleotides targeting Huntingtin HCT
  • Single Nucleotide Polymorphism SNP
  • Additional chimeric antisense oligonucleotides were designed based on 15-mer, ISIS 460209 and 17-mer, ISIS 476333 wherein the central gap region contains nine 2'-deoxynucleosides. These gapmers were designed with the central gap region shortened at the 5'-end of the the central gap region. The gapmers were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting HTT SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the gapmers were evaluated and compared to ISIS 460209 and ISIS 476333.
  • the gapmers and their motifs are described in Table 91.
  • Nucleosides without a subscript are (3-D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 or 9 as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 92.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • Table 92 a couple of the newly designed antisense oligonucleotides (ISIS 571036 and 571037) showed improvement in potency and selectivity in inhibiting mut HTT mRNA levels as compared to ISIS 460209 and 476333.
  • Table 91 Short-gap antisense oligonucleotides targeting HTT SNP ISIS NO. Sequence (5' to 3') Motif chemistry Gap Wing chemistry SEQ ID NO.
  • Short-gap chimeric oligonucleotides targeting Huntingtin HCT
  • Single Nucleotide Polymorphism SNP
  • Additional chimeric antisense oligonucleotides were designed based on 15-mer, ISIS 460209 wherein the central gap region contains nine 2'-deoxynucleosides. These gapmers were designed by having the central gap region shortened to seven 2'-deoxynucleosides. The gapmers were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting HTT SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the gapmers were evaluated and compared to ISIS 460209.
  • the gapmers and their motifs are described in Table 93.
  • Nucleosides without a subscript are ( ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 or 9 as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 94.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • each of the newly designed antisense oligonucleotides (ISIS 540108 and 571069) showed improvement in potency and/or selectivity in inhibiting mut HTT mRNA levels as compared to ISIS 460209.
  • Table 93 Short-gap antisense oligonucleotides targeting HTT SNP ISIS NO. Sequence (5' to 3') Motif Gap chemistry Wing chemistry SEQ ID NO.
  • Short-gap chimeric oligonucleotides targeting Huntingtin HCT
  • Single Nucleotide Polymorphism SNP
  • Additional chimeric antisense oligonucleotides were designed based on 15-mer, ISIS 460209 and 17-mer, ISIS 540108 wherein the central gap region contains nine and seven 2'-deoxynucleosides, respectively. These gapmers were designed by introducing one or more cEt modification(s) at the 5'-end of the central gap region. The gapmers were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting HTT SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the gapmers were evaluated and compared to ISIS 460209 and ISIS 540108.
  • the gapmers and their motifs are described in Table 95.
  • Nucleosides without a subscript are ( ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 or 9 as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 96.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • Short-gap chimeric oligonucleotides targeting Huntingtin HCT
  • Single Nucleotide Polymorphism SNP
  • Additional chimeric antisense oligonucleotides were designed based on 15-mer, ISIS 460209 and 17-mer, ISIS 540108 wherein the central gap region contains nine and seven 2'-deoxynucleosides, respectively. These gapmers were designed by introducing one or more cEt modification(s) at the 3'-end of the central gap region. The gapmers were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting HTT SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the gapmers were evaluated and compared to ISIS 460209 and ISIS 540108.
  • the gapmers and their motifs are described in Table 97.
  • Nucleosides without a subscript are ( ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicate 2'-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript "k” indicate 6'- (S) -CH 3 bicyclic nucleosides (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 or 9 as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used.
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • the HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 98.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • each of the newly designed oligonucleotides showed improvement in selective inhibition of mutant HTT mRNA levels compared to ISIS 460209. Comparable potency was observed for ISIS 568879 and 568880 while a slight loss in potency was observed for ISIS 556875, 556876 and 556877.
  • Table 97 Short-gap antisense oligonucleotides targeting HTT SNP ISIS NO. Sequence (5' to 3') Motif Gap chemistry Wing chemistry SEQ ID NO.
  • modified oligonucleotides were designed based on the parent gapmer, ISIS 460209 wherein the central gap region contains nine 2'-deoxyribonucleosides. These modified oligonucleotides were designed by introducing various chemical modifications in the central gap region and were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the modified oligonucleotides were evaluated and compared to the parent gapmer, ISIS 460209.
  • the modified oligonucleotides were created with a 3-9-3 motif and are described in Table 99.
  • Nucleosides without a subscript are ⁇ -D-2'-deoxyribonucleosides.
  • Nucleosides followed by a subscript "e” indicates a 2'-O-methoxyethyl (MOE) modified nucleoside.
  • Nucleosides followed by a subscript "k” indicates a 6'- (S) -CH 3 bicyclic nucleoside (e.g. cEt).
  • m C indicates a 5-methyl cytosine nucleoside.
  • x T indicates a 2-thio-thymidine nucleoside.
  • Underlined nucleoside indicates the position on the oligonucleotides opposite to the SNP position, which is position 8 as counted from the 5'-terminus.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used (from Coriell Institute).
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C_2229297_10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2X PCR buffer, 101 uL primers (300 uM from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN and the results are presented in Table 100.
  • the IC 50 and selectivity were calculated using methods previously described in Example 41.
  • the IC 50 at which each oligonucleotide inhibits the mutant HTT mRNA expression is denoted as 'mut IC 50 '.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as 'wt IC 50 '.
  • Selectivity was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA.
  • oligonucleotides comprising chemical modifications in the central gap region targeting Huntingtin (HTT) Single Nucleotide Polymorphism (SNP)
  • Additional chimeric antisense oligonucleotides were designed in the same manner as the antisense oligonucleotides described in Example 65. These gapmers were designed by introducing various modifications in the central gap region and were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting SNP while leaving the expression of the wild-type (wt) intact. The activity and selectivity of the modified oligonucleotides were evaluated and compared to the parent gapmer, ISIS 460209.
EP21161967.1A 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations Active EP3922722B1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP23181830.3A EP4269584A3 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201161522659P 2011-08-11 2011-08-11
US201261596723P 2012-02-08 2012-02-08
US201261603196P 2012-02-24 2012-02-24
EP12822510.9A EP2742056B2 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et utilisations de ceux-ci
PCT/US2012/050015 WO2013022984A1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et utilisations de ceux-ci
EP17150614.0A EP3205725B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP19161655.6A EP3556859B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
EP12822510.9A Division EP2742056B2 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et utilisations de ceux-ci
EP19161655.6A Division EP3556859B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP17150614.0A Division EP3205725B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP23181830.3A Division EP4269584A3 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations

Publications (2)

Publication Number Publication Date
EP3922722A1 true EP3922722A1 (fr) 2021-12-15
EP3922722B1 EP3922722B1 (fr) 2023-06-28

Family

ID=46717938

Family Applications (7)

Application Number Title Priority Date Filing Date
EP12748812.0A Active EP2742135B2 (fr) 2011-08-11 2012-08-08 Composés oligomères à brèche modifiés par liaison et leurs utilisations
EP12751399.2A Active EP2742136B1 (fr) 2011-08-11 2012-08-08 Composés oligomères à brèche comprenant des désoxyribonucléosides modifiés en 5' dans la brèche, et leurs utilisations
EP17150614.0A Active EP3205725B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP19161655.6A Active EP3556859B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP23181830.3A Pending EP4269584A3 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP21161967.1A Active EP3922722B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP12822510.9A Active EP2742056B2 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et utilisations de ceux-ci

Family Applications Before (5)

Application Number Title Priority Date Filing Date
EP12748812.0A Active EP2742135B2 (fr) 2011-08-11 2012-08-08 Composés oligomères à brèche modifiés par liaison et leurs utilisations
EP12751399.2A Active EP2742136B1 (fr) 2011-08-11 2012-08-08 Composés oligomères à brèche comprenant des désoxyribonucléosides modifiés en 5' dans la brèche, et leurs utilisations
EP17150614.0A Active EP3205725B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP19161655.6A Active EP3556859B1 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations
EP23181830.3A Pending EP4269584A3 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et leurs utilisations

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP12822510.9A Active EP2742056B2 (fr) 2011-08-11 2012-08-08 Composés antisens sélectifs et utilisations de ceux-ci

Country Status (5)

Country Link
US (9) US9752142B2 (fr)
EP (7) EP2742135B2 (fr)
DK (2) DK2742136T3 (fr)
ES (2) ES2635866T5 (fr)
WO (4) WO2013022966A1 (fr)

Families Citing this family (135)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE540118T1 (de) 2006-10-18 2012-01-15 Isis Pharmaceuticals Inc Antisense-verbindungen
BRPI0923225A2 (pt) 2008-12-02 2016-10-04 Chiralgen Ltd metodo para sintese de acidos nucleicos modificados no atomo de fosforo
MX342945B (es) 2009-07-06 2016-10-18 Ontorii Inc * Profármacos de ácido nucleico novedosos y métodos de uso de los mismos.
JP5868324B2 (ja) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan 不斉補助基
EP3467109A1 (fr) 2011-02-08 2019-04-10 Ionis Pharmaceuticals, Inc. Composés oligomères comprenant des nucléotides bicycliques et leurs utilisations
WO2013012758A1 (fr) 2011-07-19 2013-01-24 Ontorii, Inc. Procédés pour la synthèse d'acides nucléiques fonctionnalisés
EP2742135B2 (fr) 2011-08-11 2020-06-10 Ionis Pharmaceuticals, Inc. Composés oligomères à brèche modifiés par liaison et leurs utilisations
EP2839006B1 (fr) * 2012-04-20 2018-01-03 Ionis Pharmaceuticals, Inc. Composés oligomères comprenant des nucléotides bicycliques et utilisations de ceux-ci
RU2677639C2 (ru) 2012-07-13 2019-01-18 Шин Ниппон Биомедикал Лэбораториз, Лтд. Хиральный адъювант нуклеиновой кислоты
KR102450907B1 (ko) 2012-07-13 2022-10-04 웨이브 라이프 사이언시스 리미티드 키랄 제어
CN104684893B (zh) 2012-07-13 2016-10-26 日本波涛生命科学公司 不对称辅助基团
WO2014059353A2 (fr) 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Composés oligomères comportant des nucléosides bicycliques et leurs utilisations
WO2014059356A2 (fr) 2012-10-12 2014-04-17 Isis Pharmaceuticals, Inc. Composés anti-sens sélectifs et leurs utilisations
EP4144845B1 (fr) * 2012-10-12 2024-04-24 Ionis Pharmaceuticals, Inc. Composés antisens et leurs utilisations
US10260069B2 (en) 2013-02-04 2019-04-16 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
WO2014172698A1 (fr) * 2013-04-19 2014-10-23 Isis Pharmaceuticals, Inc. Compositions et procédés de modulation d'acides nucléiques par une dégradation médiée des éléments non-sens
BR112015032432B1 (pt) * 2013-06-27 2023-02-07 Roche Innovation Center Copenhagen A/S Oligômero antissenso, conjugados de oligonucleotídeo antissenso, composição farmacêutica, uso dos mesmos para o tratamento da hipercolesterolemia ou distúrbios relacionados e método in vitro para reduzir os níveis de expressão e/ou a atividade de pcsk9 em uma célula
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
JPWO2015108047A1 (ja) 2014-01-15 2017-03-23 株式会社新日本科学 免疫誘導活性を有するキラル核酸アジュバンド及び免疫誘導活性剤
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
KR20230152178A (ko) 2014-01-16 2023-11-02 웨이브 라이프 사이언시스 리미티드 키랄 디자인
WO2015168172A1 (fr) * 2014-04-28 2015-11-05 Isis Pharmaceuticals, Inc. Composés oligomères modifiés par liaison
WO2016061487A1 (fr) * 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Agents polynucléotidiques de ciblage d'acide aminolévulinique synthase-1 (alas1) et utilisations de ceux-ci
MX2017011010A (es) 2015-02-26 2017-10-20 Ionis Pharmaceuticals Inc Moduladores de rodopsina p23h con especificidad de alelo.
MA43072A (fr) 2015-07-22 2018-05-30 Wave Life Sciences Ltd Compositions d'oligonucléotides et procédés associés
US10370667B2 (en) 2015-11-18 2019-08-06 Rosalind Franklin University Of Medicine And Science Antisense compounds targeting leucine-rich repeat kinase 2 (LRRK2) for the treatment of parkinsons disease
EP3377629B1 (fr) 2015-11-18 2023-07-05 Rosalind Franklin University of Medicine and Science Composés antisens ciblant une kinase 2 à répétition riche en leucine (lrrk2) pour le traitement de la maladie de parkinson
CN113750101A (zh) 2015-12-10 2021-12-07 Ptc医疗公司 用于治疗亨廷顿病的方法
WO2017111137A1 (fr) * 2015-12-22 2017-06-29 味の素株式会社 Procédé de production d'oligonucléotides
CN107282123B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯齐聚催化剂组合物及其应用
CN107282124B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯四聚催化剂组合物及四聚方法
CN107282121B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯齐聚用催化剂组合物和齐聚方法
CN107282120B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯齐聚用催化剂组合物及齐聚方法
CN107282122B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯四聚催化剂组合物及其应用
CN107282132B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯四聚催化剂组合物及应用
CN107282130B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 乙烯四聚催化剂组合物及其应用
MA45270A (fr) * 2016-05-04 2017-11-09 Wave Life Sciences Ltd Compositions d'oligonucléotides et procédés associés
MA45496A (fr) 2016-06-17 2019-04-24 Hoffmann La Roche Molécules d'acide nucléique pour la réduction de l'arnm de padd5 ou pad7 pour le traitement d'une infection par l'hépatite b
EP3548005A4 (fr) 2016-11-29 2020-06-17 Puretech Health LLC Exosomes destinés à l'administration d'agents thérapeutiques
EP3587576A4 (fr) * 2017-02-21 2021-01-06 Osaka University Acide oligonucléique anti-sens
US11638717B2 (en) 2017-03-29 2023-05-02 Shionogi & Co., Ltd. Complex of nucleic acid medicine and multibranched lipid
SG11201911615WA (en) 2017-06-05 2020-01-30 Ptc Therapeutics Inc Compounds for treating huntington's disease
MX2019015578A (es) 2017-06-28 2020-07-28 Ptc Therapeutics Inc Metodos para tratar la enfermedad de huntington.
CA3067592A1 (fr) 2017-06-28 2019-01-03 Ptc Therapeutics, Inc. Methodes de traitement de la maladie de huntington
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use
WO2019073018A1 (fr) 2017-10-13 2019-04-18 Roche Innovation Center Copenhagen A/S Procédés d'identification de variants d'oligonucléotides phosphorothioate stéréodéfinis améliorés d'oligonucléotides antisens mettant en œuvre des sous-bibliothèques d'oligonucléotides partiellement stéréodéfinis
CR20200205A (es) 2017-10-16 2020-06-28 Hoffmann La Roche MOLECULA DE ACIDOS NUCLEICOS PARA LA REDUCCIÒN DEL ARNm DE PAPD5 Y PAPD7 EN EL TRATAMIENTO DE LA INFECCIÒN DE LA HEPATITIS B
CN111344408A (zh) 2017-12-11 2020-06-26 哥本哈根罗氏创新中心 用于调控fndc3b表达的寡核苷酸
WO2019115417A2 (fr) 2017-12-12 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucléotides pour la modulation de l'expression de rb1
EP3729095A1 (fr) 2017-12-21 2020-10-28 F. Hoffmann-La Roche AG Diagnostic compagnon pour des antagonistes d'arn de htra1
MX2020005754A (es) 2017-12-22 2020-08-20 Roche Innovation Ct Copenhagen As Oligonucleotidos gapmeros que comprenden un enlace internucleosido fosforoditioato.
KR20200104345A (ko) 2017-12-22 2020-09-03 로슈 이노베이션 센터 코펜하겐 에이/에스 포스포로디티오에이트 뉴클레오시드간 연결을 포함하는 올리고뉴클레오티드
WO2019122277A1 (fr) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Nouvelles thiophosphoramidites
US20210095274A1 (en) 2018-01-10 2021-04-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating pias4 expression
EP3737760A1 (fr) 2018-01-12 2020-11-18 Roche Innovation Center Copenhagen A/S Oligonucléotides pour la modulation de l'expression de gsk3b
CN111615558A (zh) 2018-01-17 2020-09-01 罗氏创新中心哥本哈根有限公司 用于调节erc1表达的寡核苷酸
EP3740573A1 (fr) 2018-01-18 2020-11-25 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant srebp1
WO2019145386A1 (fr) 2018-01-26 2019-08-01 Roche Innovation Center Copenhagen A/S Oligonucléotides pour la modulation de l'expression de csnk1d
CA3088071A1 (fr) 2018-02-09 2019-08-15 Genentech, Inc. Oligonucleotides pour moduler l'expression de tmem106b
JP7317029B2 (ja) 2018-02-12 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド 修飾化合物及びその使用
US20210054377A1 (en) * 2018-03-20 2021-02-25 Tokyo Institute Of Technology Antisense oligonucleotide reduced in toxicity
MX2020009957A (es) 2018-03-27 2021-01-15 Ptc Therapeutics Inc Compuestos para el tratamiento de enfermedad de hungtinton.
AU2019247645A1 (en) 2018-04-05 2020-10-15 Centre Leon Berard Use of FUBP1 inhibitors for treating hepatitis B virus infection
US20220002796A1 (en) 2018-05-07 2022-01-06 Roche Innovation Center Copenhagen A/S Quality control of lna oligonucleotide therapeutics using massively parallel sequencing
EP3790971A1 (fr) 2018-05-08 2021-03-17 Roche Innovation Center Copenhagen A/S Oligonucléotides pour moduler l'expression de myh7
JP7379387B2 (ja) 2018-06-05 2023-11-14 エフ. ホフマン-ラ ロシュ アーゲー Atxn2発現を制御するためのオリゴヌクレオチド
US20210254059A1 (en) * 2018-06-18 2021-08-19 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
PE20211378A1 (es) 2018-06-27 2021-07-27 Ptc Therapeutics Inc Compuestos heterociclicos y de heteroarilo para tratar la enfermedad de huntington
WO2020007772A1 (fr) 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant gbp-1
WO2020007700A1 (fr) 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant spi1
WO2020007702A1 (fr) 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant bcl2l11
RU2768285C1 (ru) 2018-07-03 2022-03-23 Ф. Хоффманн-Ля Рош Аг Олигонуклеотиды для модуляции экспрессии тау-белка
WO2020007889A1 (fr) 2018-07-05 2020-01-09 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant stat1
WO2020007826A1 (fr) 2018-07-05 2020-01-09 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant mbtps1
WO2020011743A1 (fr) 2018-07-09 2020-01-16 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant mafb
WO2020011653A1 (fr) 2018-07-09 2020-01-16 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant le kynu
WO2020011869A2 (fr) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant tlr2
WO2020011745A2 (fr) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant cers6
WO2020011744A2 (fr) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant cers5
KR20210033004A (ko) 2018-07-13 2021-03-25 에프. 호프만-라 로슈 아게 Rtel1의 발현을 조절하기 위한 올리고뉴클레오티드
KR20210040060A (ko) 2018-07-31 2021-04-12 로슈 이노베이션 센터 코펜하겐 에이/에스 포스포로트리티오에이트 뉴클레오시드간 연쇄를 포함하는 올리고뉴클레오티드
AU2019313443A1 (en) 2018-07-31 2020-11-26 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
WO2020038973A1 (fr) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant sptlc1
WO2020038971A1 (fr) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant la vcan
WO2020038976A1 (fr) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens ciblant l'usp8
EP3620519A1 (fr) 2018-09-04 2020-03-11 F. Hoffmann-La Roche AG Utilisation de vésicules extracellulaires de lait isolées pour l'administration orale d'oligonucléotides
CN112969709A (zh) 2018-11-01 2021-06-15 豪夫迈·罗氏有限公司 靶向tia1的反义寡核苷酸
WO2020109343A1 (fr) 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Polythérapie pour le traitement de la dégénérescence maculaire
WO2020109344A1 (fr) 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Dispositif d'administration oculaire pour oligonucléotides antisens
US20220042011A1 (en) 2018-12-21 2022-02-10 Hoffmann-La Roche Inc. Antisense oligonucleotides targeting card9
WO2020152303A1 (fr) 2019-01-25 2020-07-30 F. Hoffmann-La Roche Ag Vésicule lipidique pour administration de médicament par voie orale
US20220098595A1 (en) * 2019-01-25 2022-03-31 Nayan Therapeutics, Inc. Nr2e3 expression reducing oligonucleotides, compositions containing the same, and methods of their use
US20220162606A1 (en) * 2019-01-25 2022-05-26 Nayan Therapeutics, Inc. Nrl expression reducing oligonucleotides, compositions containing the same, and methods of their use
AU2020216186A1 (en) * 2019-02-01 2021-07-29 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
WO2020169695A1 (fr) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S Oligonucléotides gapmer de phosphonoacétate
MX2021009949A (es) 2019-02-20 2021-09-21 Roche Innovation Ct Copenhagen As Fosforamiditas novedosas.
CN113474633A (zh) 2019-02-26 2021-10-01 罗氏创新中心哥本哈根有限公司 寡核苷酸配制方法
US11286485B2 (en) 2019-04-04 2022-03-29 Hoffmann-La Roche Inc. Oligonucleotides for modulating ATXN2 expression
MX2021012981A (es) * 2019-04-25 2022-01-18 Wave Life Sciences Ltd Composiciones de oligonucleotido y metodos de uso de las mismas.
JP7155302B2 (ja) 2019-06-06 2022-10-18 エフ.ホフマン-ラ ロシュ アーゲー Atxn3を標的とするアンチセンスオリゴヌクレオチド
WO2021021673A1 (fr) * 2019-07-26 2021-02-04 Ionis Pharmaceuticals, Inc. Composés et procédés pour la modulation de gfap
JP2022544587A (ja) * 2019-08-15 2022-10-19 アイオーニス ファーマシューティカルズ, インコーポレーテッド 結合修飾オリゴマー化合物及びその使用
CA3156848A1 (fr) 2019-11-01 2021-05-06 Novartis Ag Utilisation d'un modulateur d'epissage pour un traitement ralentissant la progression de la maladie de huntington
EP4077667A1 (fr) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Utilisation d'inhibiteurs de sept9 pour traiter une infection par le virus de l'hépatite b
WO2021122910A1 (fr) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Utilisation d'inhibiteurs de sbds pour traiter une infection par le virus de l'hépatite b
CN114829599A (zh) 2019-12-19 2022-07-29 豪夫迈·罗氏有限公司 Scamp3抑制剂用于治疗乙型肝炎病毒感染的用途
EP4077670A1 (fr) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Utilisation d'inhibiteurs de cops3 pour traiter une infection par le virus de l'hépatite b
CN114867856A (zh) 2019-12-19 2022-08-05 豪夫迈·罗氏有限公司 Saraf抑制剂用于治疗乙型肝炎病毒感染的用途
US20210214727A1 (en) 2019-12-20 2021-07-15 Hoffmann-La Roche Inc. Enhanced oligonucleotides for inhibiting scn9a expression
CN114828852A (zh) 2019-12-24 2022-07-29 豪夫迈·罗氏有限公司 用于治疗hbv的靶向hbv的抗病毒药剂和/或免疫调节剂的药物组合
EP4081639A1 (fr) 2019-12-24 2022-11-02 F. Hoffmann-La Roche AG Association pharmaceutique d'un oligonucléotide thérapeutique ciblant le vhb et un agoniste de tlr7 pour le traitement du vhb
CA3161513A1 (fr) 2020-01-28 2021-08-05 Irwin DAVIDSON Oligonucleotide antisens ciblant linc00518 pour le traitement du melanome
EP4110916A1 (fr) 2020-02-28 2023-01-04 F. Hoffmann-La Roche AG Oligonucléotides pour moduler l'épissage de l'exon 7 de cd73
JP2023527693A (ja) 2020-05-11 2023-06-30 ジェネンテック, インコーポレイテッド 神経疾患を治療するための補体成分c1r阻害剤、並びに関連する組成物、システム、及びそれを使用する方法
WO2021231204A1 (fr) 2020-05-11 2021-11-18 Genentech, Inc. Inhibiteurs du composant 4 du complément pour le traitement de maladies neurologiques, et compositions associées, systèmes et procédés d'utilisation de ceux-ci
EP4149486A1 (fr) 2020-05-11 2023-03-22 Genentech, Inc. Inhibiteurs de la composante c1s du complément pour le traitement d'une maladie neurologique, et compositions associées, systèmes et procédés d'utilisation de ceux-ci
JP2023526096A (ja) 2020-05-22 2023-06-20 エフ. ホフマン-ラ ロシュ アーゲー Card9のスプライス調節のためのオリゴヌクレオチド
US20230212572A1 (en) 2020-06-09 2023-07-06 Roche Innovation Center Copenhagen A/S Guanosine Analogues for Use in Therapeutics Polynucleotides
AR122731A1 (es) 2020-06-26 2022-10-05 Hoffmann La Roche Oligonucleótidos mejorados para modular la expresión de fubp1
WO2022018155A1 (fr) 2020-07-23 2022-01-27 F. Hoffmann-La Roche Ag Oligonucléotides lna pour la modulation d'épissage de stmn2
JP2023538630A (ja) 2020-08-21 2023-09-08 エフ. ホフマン-ラ ロシュ アーゲー B型肝炎ウイルス感染症を処置するためのa1cf阻害剤の使用
EP4136092A4 (fr) 2020-11-18 2023-10-11 Ionis Pharmaceuticals, Inc. Composés et procédés pour moduler l'expression de l'angiotensinogène
AR124227A1 (es) 2020-12-03 2023-03-01 Hoffmann La Roche Oligonucleótidos antisentido que actúan sobre atxn3
AR124229A1 (es) 2020-12-03 2023-03-01 Hoffmann La Roche Oligonucleótidos antisentido que actúan sobre atxn3
EP4259642A1 (fr) 2020-12-08 2023-10-18 F. Hoffmann-La Roche AG Nouvelle synthèse d'oligonucléotides phosphorodithioate
WO2022162015A1 (fr) 2021-01-26 2022-08-04 Universite Brest Bretagne Occidentale Nouveaux variants d'épissage de stim1 et leurs utilisations
TW202246500A (zh) 2021-02-02 2022-12-01 瑞士商赫孚孟拉羅股份公司 用於抑制 rtel1 表現之增強型寡核苷酸
TW202304446A (zh) 2021-03-29 2023-02-01 瑞士商諾華公司 剪接調節子用於減慢杭丁頓氏舞蹈症進展的治療之用途
WO2023052317A1 (fr) 2021-09-29 2023-04-06 F. Hoffmann-La Roche Ag Édition d'arn
WO2023078883A1 (fr) 2021-11-03 2023-05-11 F. Hoffmann-La Roche Ag Oligonucléotides pour moduler l'expression de l'apolipoprotéine e4
WO2023083906A2 (fr) 2021-11-11 2023-05-19 F. Hoffmann-La Roche Ag Combinaisons pharmaceutiques pour le traitement du vhb
WO2023111210A1 (fr) 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Combinaison d'oligonucléotides pour moduler rtel1 et fubp1
WO2023117738A1 (fr) 2021-12-20 2023-06-29 F. Hoffmann-La Roche Ag Oligonucléotides antisens d'acide nucléique à thréose et procédés associés
WO2023141507A1 (fr) 2022-01-20 2023-07-27 Genentech, Inc. Oligonucléotides antisens pour moduler l'expression de tmem106b
EP4332221A1 (fr) 2022-08-29 2024-03-06 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens d'acide nucléique à thréose et procédés associés

Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4845205A (en) 1985-01-08 1989-07-04 Institut Pasteur 2,N6 -disubstituted and 2,N6 -trisubstituted adenosine-3'-phosphoramidites
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
WO1994014226A1 (fr) 1992-12-14 1994-06-23 Honeywell Inc. Systeme de moteur a tolerance de pannes
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5552540A (en) 1987-06-24 1996-09-03 Howard Florey Institute Of Experimental Physiology And Medicine Nucleoside derivatives
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5614617A (en) 1990-07-27 1997-03-25 Isis Pharmaceuticals, Inc. Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5645985A (en) 1991-11-26 1997-07-08 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5750692A (en) 1990-01-11 1998-05-12 Isis Pharmaceuticals, Inc. Synthesis of 3-deazapurines
US5830653A (en) 1991-11-26 1998-11-03 Gilead Sciences, Inc. Methods of using oligomers containing modified pyrimidines
US6268490B1 (en) 1997-03-07 2001-07-31 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogues
US6525191B1 (en) 1999-05-11 2003-02-25 Kanda S. Ramasamy Conformationally constrained L-nucleosides
US6670461B1 (en) 1997-09-12 2003-12-30 Exiqon A/S Oligonucleotide analogues
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US20040171570A1 (en) 2002-11-05 2004-09-02 Charles Allerson Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004106356A1 (fr) 2003-05-27 2004-12-09 Syddansk Universitet Derives de nucleotides fonctionnalises
WO2005021570A1 (fr) 2003-08-28 2005-03-10 Gene Design, Inc. Nouveaux acides nucleiques artificiels de type a liaison n-o reticulee
US20050130923A1 (en) 2003-09-18 2005-06-16 Balkrishen Bhat 4'-thionucleosides and oligomeric compounds
US7053207B2 (en) 1999-05-04 2006-05-30 Exiqon A/S L-ribo-LNA analogues
WO2007134181A2 (fr) 2006-05-11 2007-11-22 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques bicycliques modifiés en 5'
US20080039618A1 (en) 2002-11-05 2008-02-14 Charles Allerson Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
US7399845B2 (en) 2006-01-27 2008-07-15 Isis Pharmaceuticals, Inc. 6-modified bicyclic nucleic acid analogs
WO2008101157A1 (fr) 2007-02-15 2008-08-21 Isis Pharmaceuticals, Inc. Nucléosides modifiés 5'-substitués-2'-f et composés oligomères préparés à partir de ceux-ci
WO2008150729A2 (fr) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques bicycliques pontés par aminométhylène n-substitué
WO2008154401A2 (fr) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Analogues d'acide nucléique bicyclique carbocylique
WO2009006478A2 (fr) 2007-07-05 2009-01-08 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques bicycliques disubstitués en position 6
WO2009061851A2 (fr) 2007-11-09 2009-05-14 Isis Pharmaceuticals, Inc. Modulation de l'expression du facteur 7
WO2009135322A1 (fr) 2008-05-09 2009-11-12 The Universtity Of British Columbia Procédés et compositions pour le traitement de la maladie de huntington

Family Cites Families (161)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2699808A (en) 1944-10-06 1955-01-18 Mark W Lowe Apparatus for peeling tomatoes
US2699508A (en) 1951-12-21 1955-01-11 Selectronics Inc Method of mounting and construction of mounting for low frequency piezoelectric crystals
JPS5717316A (en) 1980-07-04 1982-01-29 Kawasaki Steel Corp Method for automatic control of screw down of reeler mill
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
FR2567892B1 (fr) 1984-07-19 1989-02-17 Centre Nat Rech Scient Nouveaux oligonucleotides, leur procede de preparation et leurs applications comme mediateurs dans le developpement des effets des interferons
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
ES2116977T3 (es) 1990-05-11 1998-08-01 Microprobe Corp Soportes solidos para ensayos de hibridacion de acidos nucleicos y metodos para inmovilizar oligonucleotidos de modo covalente.
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5965722A (en) 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
EP0538194B1 (fr) 1991-10-17 1997-06-04 Novartis AG Nucléosides et oligonucléosides bicycliques, leur procédé de préparation et leurs intermédiaires
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US20010044145A1 (en) 1991-12-24 2001-11-22 Monia Brett P. Methods of using mammalian RNase H and compositions thereof
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
FR2687679B1 (fr) 1992-02-05 1994-10-28 Centre Nat Rech Scient Oligothionucleotides.
FR2692265B1 (fr) 1992-05-25 1996-11-08 Centre Nat Rech Scient Composes biologiquement actifs de type phosphotriesters.
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (fr) 1992-07-01 1994-01-05 Ciba-Geigy Ag Nucléosides carbocycliques contenant des noyaux bicycliques, oligonucléotides en dérivant, procédé pour leur préparation, leur application et des intermédiaires
EP0652890B1 (fr) 1992-07-27 1998-01-14 HYBRIDON, Inc. Alkylphosphonothioates oligonucleotidiques
CN1121721A (zh) 1993-01-25 1996-05-01 海布里顿公司 寡核苷酸烷基膦酸酯和烷基硫代膦酸酯
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
US5446786A (en) 1993-03-03 1995-08-29 Northern Telecom Limited Two-wire telecommunications line detection arrangements
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
EP0691968B1 (fr) 1993-03-30 1997-07-16 Sanofi Analogues de nucleosides acycliques et sequences d'oligonucleotides contenant ceux-ci
AU6449294A (en) 1993-03-31 1994-10-24 Sterling Winthrop Inc. Novel 5'-substituted nucleosides and oligomers produced therefrom
DE4311944A1 (de) 1993-04-10 1994-10-13 Degussa Umhüllte Natriumpercarbonatpartikel, Verfahren zu deren Herstellung und sie enthaltende Wasch-, Reinigungs- und Bleichmittelzusammensetzungen
FR2705099B1 (fr) 1993-05-12 1995-08-04 Centre Nat Rech Scient Oligonucléotides phosphorothioates triesters et procédé de préparation.
US5801154A (en) 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
WO1995014030A1 (fr) * 1993-11-16 1995-05-26 Genta Incorporated Oligomeres synthetiques ayant des liaisons internucleosidyle phosphonate chiralement pures melangees avec des liaisons internucleosidyle non phosphonate
NZ277617A (en) * 1993-11-16 1998-01-26 Genta Inc Chimeric oligonucleoside compounds useful in activating rnaseh mediated cleavage of ribonucleic acid sequences
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5595756A (en) 1993-12-22 1997-01-21 Inex Pharmaceuticals Corporation Liposomal compositions for enhanced retention of bioactive agents
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5646269A (en) 1994-04-28 1997-07-08 Gilead Sciences, Inc. Method for oligonucleotide analog synthesis
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5792747A (en) 1995-01-24 1998-08-11 The Administrators Of The Tulane Educational Fund Highly potent agonists of growth hormone releasing hormone
US20050054600A1 (en) * 1995-08-17 2005-03-10 Hybridon, Inc. Modified protein kinase a-specific oligonucleotides and methods of their use
US6624293B1 (en) * 1995-08-17 2003-09-23 Hybridon, Inc. Modified protein kinase A-specific oligonucleotides and methods of their use
US6331617B1 (en) * 1996-03-21 2001-12-18 University Of Iowa Research Foundation Positively charged oligonucleotides as regulators of gene expression
US5656408A (en) 1996-04-29 1997-08-12 Xerox Corporation Coated carrier particles
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
ES2192672T3 (es) 1996-11-18 2003-10-16 Takeshi Imanishi Nuevos analogos de nucleotidos.
DE69829760T3 (de) 1997-09-12 2016-04-14 Exiqon A/S Bi- und tri-zyklische - nukleosid, nukleotid und oligonukleotid-analoga
US20030228597A1 (en) 1998-04-13 2003-12-11 Cowsert Lex M. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
CN1273478C (zh) 1999-02-12 2006-09-06 三共株式会社 新型核苷及低聚核苷酸类似物
US6753422B2 (en) 1999-03-01 2004-06-22 O'brien Thomas G. Odc allelic analysis method for assessing carcinogenic susceptibility
CA2363077A1 (fr) 1999-03-01 2000-09-08 Variagenics, Inc. Methodes permettant de cibler des molecules d'arn
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US5998148A (en) 1999-04-08 1999-12-07 Isis Pharmaceuticals Inc. Antisense modulation of microtubule-associated protein 4 expression
JP4151751B2 (ja) 1999-07-22 2008-09-17 第一三共株式会社 新規ビシクロヌクレオシド類縁体
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US20020187931A1 (en) 2000-04-13 2002-12-12 Michael Hayden Modulating cell survival by modulating huntingtin function
US7135565B2 (en) 2000-07-28 2006-11-14 Agilent Technologies, Inc. Synthesis of polynucleotides using combined oxidation/deprotection chemistry
WO2002027033A1 (fr) 2000-09-29 2002-04-04 Isis Pharmaceuticals, Inc. Modulation anti-sens de l'expression de mekk4
US6693187B1 (en) 2000-10-17 2004-02-17 Lievre Cornu Llc Phosphinoamidite carboxlates and analogs thereof in the synthesis of oligonucleotides having reduced internucleotide charge
US6426220B1 (en) 2000-10-30 2002-07-30 Isis Pharmaceuticals, Inc. Antisense modulation of calreticulin expression
US20050191638A1 (en) 2002-02-20 2005-09-01 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
WO2002094250A2 (fr) 2001-05-18 2002-11-28 Cureon A/S Utilisations therapeutiques d'oligonucleotides modifies par lna dans des maladies infectieuses
WO2003004602A2 (fr) 2001-07-03 2003-01-16 Isis Pharmaceuticals, Inc. Oligonucleotides chimeres resistants a la nuclease
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US7888324B2 (en) 2001-08-01 2011-02-15 Genzyme Corporation Antisense modulation of apolipoprotein B expression
US20040096880A1 (en) 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
EP1423537A4 (fr) 2001-08-07 2006-11-29 Univ Delaware Compositions et procedes de prevention et traitement de la maladie d'huntington
PT1470144E (pt) * 2002-02-01 2009-02-10 Univ Mcgill Oligonucleótidos incluindo segmentos alternantes e as suas utilizações
EP2213737B1 (fr) 2002-02-01 2012-11-07 Life Technologies Corporation Oligonucleotides double brin
US20050096284A1 (en) 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
JP4338527B2 (ja) 2002-04-05 2009-10-07 サンタリス ファーマ アー/エス HIF−1α発現を調節するオリゴマー化合物
US7569575B2 (en) 2002-05-08 2009-08-04 Santaris Pharma A/S Synthesis of locked nucleic acid derivatives
US20040092465A1 (en) 2002-11-11 2004-05-13 Isis Pharmaceuticals Inc. Modulation of huntingtin interacting protein 1 expression
US20040102398A1 (en) 2002-11-23 2004-05-27 Isis Pharmaceuticals Inc. Modulation of B7H expression
US20050106731A1 (en) 2002-08-05 2005-05-19 Davidson Beverly L. siRNA-mediated gene silencing with viral vectors
US20050255086A1 (en) 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20080274989A1 (en) 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
EP1546344A4 (fr) 2002-09-18 2007-10-03 Isis Pharmaceuticals Inc Reduction efficace d'arn cibles au moyen de composes oligomeres a brin simple et double
US20040219565A1 (en) 2002-10-21 2004-11-04 Sakari Kauppinen Oligonucleotides useful for detecting and analyzing nucleic acids of interest
EP1569695B1 (fr) 2002-11-13 2013-05-15 Genzyme Corporation Modulation antisens de l'expression d'apolipoproteine b
EP2305813A3 (fr) 2002-11-14 2012-03-28 Dharmacon, Inc. SIRNA fonctionnel et hyperfonctionnel
CA2506576C (fr) 2002-11-18 2018-03-06 Santaris Pharma A/S Oligonucleotides antisens gapmeres
JP4503590B2 (ja) 2003-02-10 2010-07-14 サンタリス ファーマ アー/エス サバイビン発現を調整するためのオリゴマー化合物
CA2526893C (fr) 2003-05-14 2010-10-26 Japan Science And Technology Agency Inhibition de l'expression du gene de la huntingtine
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US20050074801A1 (en) 2003-09-09 2005-04-07 Monia Brett P. Chimeric oligomeric compounds comprising alternating regions of northern and southern conformational geometry
ES2485848T3 (es) 2003-09-12 2014-08-14 University Of Massachusetts ARN de interferencia para el tratamiento de trastornos relacionados con la ganancia de función
AR045937A1 (es) 2003-09-18 2005-11-16 Lilly Co Eli Modulacion de la expresion del factor de iniciacion eucariotico eif4e
WO2005045032A2 (fr) 2003-10-20 2005-05-19 Sima Therapeutics, Inc. Inhibition mediee par une interference arn de l'expression du gene de reponse de croissance precoce, au moyen d'un petit acide nucleique interferant (sina)
DK1706489T3 (da) 2003-12-23 2010-09-13 Santaris Pharma As Oligomer forbindelser for modulationen af BCL-2
US20050176045A1 (en) 2004-02-06 2005-08-11 Dharmacon, Inc. SNP discriminatory siRNA
GB0407382D0 (en) 2004-03-31 2004-05-05 Univ Cambridge Tech Therapeutic methods and means
WO2005105995A2 (fr) 2004-04-14 2005-11-10 Sirna Therapeutics, Inc. Traitement de maladies a expansion de sequence repetee de polyglutamine (polyq) a mediation d'arn interferent mettant en oeuvre un acide nucleique court interferent (sina)
WO2005116204A1 (fr) 2004-05-11 2005-12-08 Rnai Co., Ltd. Polynucléotide provoquant l'interférence rna et procédé de regulation d'expression génétique avec l’usage de ce dernier
AU2005252663B2 (en) 2004-06-03 2011-07-07 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US7323308B2 (en) 2004-09-03 2008-01-29 Affymetrix, Inc. Methods of genetic analysis of E. coli
EP2397563A3 (fr) 2004-09-17 2012-07-18 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
WO2007002904A2 (fr) 2005-06-28 2007-01-04 Medtronic, Inc. Procedes et sequences permettant de supprimer de maniere preferentielle l'expression de la huntingtine mutee
EP2338992A3 (fr) * 2005-08-29 2011-10-12 Regulus Therapeutics, Inc Composes antisens ayant une activite anti-microarn amelioree
JP5523705B2 (ja) 2005-08-29 2014-06-18 レグルス・セラピューティクス・インコーポレイテッド Mir−122aをモジュレートする使用方法
EP2325315B1 (fr) 2005-10-28 2014-05-07 Alnylam Pharmaceuticals, Inc. Compositions et procédés d'inhibition d'expression du gène huntingtin
JP5425474B2 (ja) 2006-01-26 2014-02-26 アイシス ファーマシューティカルズ, インコーポレーテッド ハンチンチン対する、組成物及びその使用
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
US8935416B2 (en) 2006-04-21 2015-01-13 Fortinet, Inc. Method, apparatus, signals and medium for enforcing compliance with a policy on a client computer
EP2023939B1 (fr) 2006-05-05 2012-06-27 Isis Pharmaceuticals, Inc. Composes et procedes de modulation de l'expression de pcsk9
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
US9273356B2 (en) 2006-05-24 2016-03-01 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
CA2662704A1 (fr) 2006-07-07 2008-01-10 University Of Massachusetts Compositions de silencage de l'arn, et methodes de traitement de la choree de huntington
JP4798093B2 (ja) 2006-08-04 2011-10-19 日産自動車株式会社 流体改質装置及びこれを用いた流体改質方法
AU2007282224B2 (en) 2006-08-11 2013-08-29 Vico Therapeutics B.V. Methods and means for treating DNA repeat instability associated genetic disorders
ATE540118T1 (de) 2006-10-18 2012-01-15 Isis Pharmaceuticals Inc Antisense-verbindungen
EP2102340A2 (fr) 2006-11-27 2009-09-23 Isis Pharmaceuticals, Inc. Procédés pour traiter l'hypercholestérolémie
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
EP2014769B1 (fr) 2007-06-18 2010-03-31 Commissariat à l'Energie Atomique Amortissement du bruit à base de sirna réversible de gène huntingtin de type sauvage endogènes et mutés et son application pour le traitement de la maladie de Hungtington
US8088904B2 (en) 2007-08-15 2012-01-03 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
WO2009067647A1 (fr) 2007-11-21 2009-05-28 Isis Pharmaceuticals, Inc. Analogues d'acide nucléique alpha-l-bicyclique carbocyclique
WO2009100320A2 (fr) 2008-02-07 2009-08-13 Isis Pharmaceuticals, Inc. Analogues d’acides nucléiques de cyclohexitol bicycliques
WO2009124238A1 (fr) * 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Composés oligomères comprenant des nucléosides bicycliques terminaux liés de façon neutre
US8846639B2 (en) * 2008-04-04 2014-09-30 Isis Pharmaceutical, Inc. Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
US8679750B2 (en) 2008-05-09 2014-03-25 The University Of British Columbia Methods and compositions for the treatment of Huntington'S disease
WO2009143369A2 (fr) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc. Procédé de préparation de nucléosides et de leurs analogues sans utiliser de chromatographie
EP2356129B1 (fr) 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Nucléosides alpha-l-bicycliques substitués
EP2447274B1 (fr) 2008-10-24 2017-10-04 Ionis Pharmaceuticals, Inc. Composants oligomères et procédés
AT507215B1 (de) 2009-01-14 2010-03-15 Boehler Edelstahl Gmbh & Co Kg Verschleissbeständiger werkstoff
EP2462153B1 (fr) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques cyclohexoses bicycliques
US9574191B2 (en) 2010-02-03 2017-02-21 The Board Of Regents Of The University Of Texas System Selective inhibition of polyglutamine protein expression
EP3628750A1 (fr) * 2010-02-08 2020-04-01 Ionis Pharmaceuticals, Inc. Réduction sélective de variants alléliques
CA2789005A1 (fr) * 2010-02-08 2011-08-11 Isis Pharmaceuticals, Inc. Reduction selective de variants alleliques
WO2011115818A1 (fr) 2010-03-17 2011-09-22 Isis Pharmaceuticals, Inc. Nucléosides bicycliques 5'-substitués et composés oligomères synthétisés à partir desdits nucléosides
CN103154014B (zh) 2010-04-28 2015-03-25 Isis制药公司 修饰核苷、其类似物以及由它们制备的寡聚化合物
EP3467109A1 (fr) * 2011-02-08 2019-04-10 Ionis Pharmaceuticals, Inc. Composés oligomères comprenant des nucléotides bicycliques et leurs utilisations
WO2012141960A1 (fr) 2011-04-11 2012-10-18 Boston Scientific Neuromodulation Corporation Systèmes et procédés pour améliorer le placement d'une dérivation de palette
DK3505528T3 (da) 2011-04-21 2021-02-08 Ionis Pharmaceuticals Inc Modulation af hepatitis-b-virus (hbv)-ekspression
EP2742135B2 (fr) 2011-08-11 2020-06-10 Ionis Pharmaceuticals, Inc. Composés oligomères à brèche modifiés par liaison et leurs utilisations
US9778706B2 (en) 2012-02-24 2017-10-03 Blackberry Limited Peekable user interface on a portable electronic device
EP2839006B1 (fr) * 2012-04-20 2018-01-03 Ionis Pharmaceuticals, Inc. Composés oligomères comprenant des nucléotides bicycliques et utilisations de ceux-ci
US9400902B2 (en) 2012-05-22 2016-07-26 Trimble Navigation Limited Multi-modal entity tracking and display
US9984408B1 (en) 2012-05-30 2018-05-29 Amazon Technologies, Inc. Method, medium, and system for live video cooperative shopping
WO2014059353A2 (fr) 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Composés oligomères comportant des nucléosides bicycliques et leurs utilisations
WO2014059356A2 (fr) 2012-10-12 2014-04-17 Isis Pharmaceuticals, Inc. Composés anti-sens sélectifs et leurs utilisations
US10260069B2 (en) 2013-02-04 2019-04-16 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US9778708B1 (en) 2016-07-18 2017-10-03 Lenovo Enterprise Solutions (Singapore) Pte. Ltd. Dual sided latching retainer for computer modules
JP7317029B2 (ja) * 2018-02-12 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド 修飾化合物及びその使用

Patent Citations (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US4845205A (en) 1985-01-08 1989-07-04 Institut Pasteur 2,N6 -disubstituted and 2,N6 -trisubstituted adenosine-3'-phosphoramidites
US5552540A (en) 1987-06-24 1996-09-03 Howard Florey Institute Of Experimental Physiology And Medicine Nucleoside derivatives
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5750692A (en) 1990-01-11 1998-05-12 Isis Pharmaceuticals, Inc. Synthesis of 3-deazapurines
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587469A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides containing N-2 substituted purines
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5614617A (en) 1990-07-27 1997-03-25 Isis Pharmaceuticals, Inc. Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5830653A (en) 1991-11-26 1998-11-03 Gilead Sciences, Inc. Methods of using oligomers containing modified pyrimidines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5645985A (en) 1991-11-26 1997-07-08 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
WO1994014226A1 (fr) 1992-12-14 1994-06-23 Honeywell Inc. Systeme de moteur a tolerance de pannes
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5763588A (en) 1993-09-17 1998-06-09 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US6005096A (en) 1993-09-17 1999-12-21 Gilead Sciences, Inc. Pyrimidine derivatives
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US6268490B1 (en) 1997-03-07 2001-07-31 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogues
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US7034133B2 (en) 1997-09-12 2006-04-25 Exiqon A/S Oligonucleotide analogues
US6670461B1 (en) 1997-09-12 2003-12-30 Exiqon A/S Oligonucleotide analogues
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US7053207B2 (en) 1999-05-04 2006-05-30 Exiqon A/S L-ribo-LNA analogues
US6525191B1 (en) 1999-05-11 2003-02-25 Kanda S. Ramasamy Conformationally constrained L-nucleosides
US20080039618A1 (en) 2002-11-05 2008-02-14 Charles Allerson Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
US20040171570A1 (en) 2002-11-05 2004-09-02 Charles Allerson Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004106356A1 (fr) 2003-05-27 2004-12-09 Syddansk Universitet Derives de nucleotides fonctionnalises
WO2005021570A1 (fr) 2003-08-28 2005-03-10 Gene Design, Inc. Nouveaux acides nucleiques artificiels de type a liaison n-o reticulee
US7427672B2 (en) 2003-08-28 2008-09-23 Takeshi Imanishi Artificial nucleic acids of n-o bond crosslinkage type
US20050130923A1 (en) 2003-09-18 2005-06-16 Balkrishen Bhat 4'-thionucleosides and oligomeric compounds
US7399845B2 (en) 2006-01-27 2008-07-15 Isis Pharmaceuticals, Inc. 6-modified bicyclic nucleic acid analogs
WO2007134181A2 (fr) 2006-05-11 2007-11-22 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques bicycliques modifiés en 5'
US20070287831A1 (en) 2006-05-11 2007-12-13 Isis Pharmaceuticals, Inc 5'-modified bicyclic nucleic acid analogs
WO2008101157A1 (fr) 2007-02-15 2008-08-21 Isis Pharmaceuticals, Inc. Nucléosides modifiés 5'-substitués-2'-f et composés oligomères préparés à partir de ceux-ci
WO2008150729A2 (fr) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques bicycliques pontés par aminométhylène n-substitué
WO2008154401A2 (fr) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Analogues d'acide nucléique bicyclique carbocylique
WO2009006478A2 (fr) 2007-07-05 2009-01-08 Isis Pharmaceuticals, Inc. Analogues d'acides nucléiques bicycliques disubstitués en position 6
WO2009061851A2 (fr) 2007-11-09 2009-05-14 Isis Pharmaceuticals, Inc. Modulation de l'expression du facteur 7
WO2009135322A1 (fr) 2008-05-09 2009-11-12 The Universtity Of British Columbia Procédés et compositions pour le traitement de la maladie de huntington

Non-Patent Citations (79)

* Cited by examiner, † Cited by third party
Title
"GENBANK", Database accession no. NT_006081.18
"Remington's Pharmaceutical Sciences", 2005, MACK PUBLISHING
ALBAEK ET AL., J. ORG. CHEM., vol. 71, 2006, pages 7731 - 7740
AM. J. HUM. GENET, vol. 81, 2007, pages 596
AM. J. HUM. GENET., vol. 78, 2006, pages 815
BRAASCH ET AL., CHEM. BIOL., vol. 8, 2001, pages 1 - 7
CARDIOLOGY, vol. 100, 2003, pages 109
CELL, vol. 130, 2007, pages 427
CHATTOPADHYAYA ET AL., J. ORG. CHEM., vol. 74, 2009, pages 118 - 134
CIRCULATION, vol. 108, 2003, pages 2971
CNS NEUROL. DISORD. DRUG TARGETS, vol. 5, 2006, pages 167
CROOKE ET AL., J. PHARMACOL. EXP. THER., vol. 277, 1996, pages 923 - 937
DE MESMAEKER ALAIN ET AL: "Amide Backbones with Conformationally Restricted Furanose Rings: Highly Improved Affinity of the Modified Oligonucleotides for Their RNA Complements", ANGEWANDTE CHEMIE - INTERNATIONAL EDITION, vol. 35, no. 23/24, 1 December 1996 (1996-12-01), pages 2790 - 2794, XP055857349 *
ELAYADI ET AL., CURR. OPINION INVENS. DRUGS, vol. 2, 2001, pages 558 - 561
ENA, MORITA ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 12, 2002, pages 73 - 76
ENGLISCH ET AL., ANGEWANDTE CHEMIE, vol. 30, 1991, pages 613
EUR. RESPIR. J., vol. 32, 2008, pages 327
FREIER ET AL., NUCLEIC ACIDS RESEARCH, vol. 25, no. 22, 1997, pages 4429 - 4443
FRIEDEN ET AL., NUCLEIC ACIDS RESEARCH, vol. 21, 2003, pages 6365 - 6372
GALLO ET AL., TETRAHEDRON, vol. 57, 2001, pages 5707 - 5713
GOUSSET HERVÉ ET AL: "Conformational Study of DNA-RNA Duplexes Containing MMI Substituted Phosphodiester Linkages by FTIR Spectroscopy", JOURNAL OF BIOMOLECULAR STRUCTURE & DYNAMICS, vol. 15, no. 5, 1 April 1998 (1998-04-01), US, pages 931 - 936, XP055857599, ISSN: 0739-1102, DOI: 10.1080/07391102.1998.10508213 *
GUILLERM ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 5, 1995, pages 1455 - 1460
HARRY-O'KURU ET AL., J. ORG. CHEM., vol. 62, no. 6, 1997, pages 1754 - 1759
HU J ET AL: "Allele-selective inhibition of mutant huntingtin by peptide nucleic acid-peptide conjugates, locked nucleic acid, and small interfering RNA", ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, NEW YORK ACADEMY OF SCIENCES, US, vol. 1175, 1 September 2009 (2009-09-01), pages 24 - 31, XP009146688, ISSN: 0077-8923, [retrieved on 20090924], DOI: 10.1111/J.1749-6632.2009.04975.X *
HUM MUTAT., vol. 30, 2009, pages 520
HUM. MOL. GEN., vol. 13, 2004, pages 171
HUM. MOL. GENE.T, vol. 19, 2010, pages 671
J STEROID BIOCHEM. MOL. BIOL., vol. 108, 2008, pages 245
J. CLIN. ENDOCRINOL. METAB., vol. 88, 2003, pages 4911
J. CLIN. INVEST., vol. 111, 2003, pages 145
J. NEUROSCI., vol. 26, 2006, pages 111623
JACOBSON ET AL., J. MED. CHEM. LETT, vol. 43, 2000, pages 2196 - 2203
KABANOV ET AL., FEBS LETT, vol. 259, 1990, pages 327 - 330
KAWASAKI ET AL., J. MED. CHEM., vol. 36, 1993, pages 831 - 841
KEITH T. GAGNON ET AL: "Allele-Selective Inhibition of Mutant Huntingtin Expression with Antisense Oligonucleotides Targeting the Expanded CAG Repeat", BIOCHEMISTRY, vol. 49, no. 47, 30 November 2010 (2010-11-30), pages 10166 - 10178, XP055033274, ISSN: 0006-2960, DOI: 10.1021/bi101208k *
KIDNEY INT., vol. 71, 2007, pages 1155
KOSHKIN ET AL., TETRAHEDRON, vol. 54, 1998, pages 3607 - 3630
KUMAR ET AL., BIOORG. MED. CHEM. LETT., vol. 8, 1998, pages 2219 - 2222
LEE ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 11, 2001, pages 1333 - 1337
LETSINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 6553 - 6556
LEUMANN, CJ, BIOORG. &MED. CHEM., vol. 10, 2002, pages 841 - 854
LEUMANN, J. C, BIOORGANIC & MEDICINAL CHEMISTRY, vol. 10, 2002, pages 841 - 854
LNA, SINGH ET AL., CHEM. COMMUN., vol. 4, 1998, pages 455 - 456
MANOHARAN ET AL., ANN. N.Y. ACAD. SCI., vol. 660, 1992, pages 306 - 309
MANOHARAN ET AL., BIOORG. MED. CHEM. LET., vol. 3, 1993, pages 2765 - 2770
MANOHARAN ET AL., BIOORG. MED. CHEM. LET., vol. 4, no. 10, 1994, pages 53 - 1060
MANOHARAN ET AL., NUCLEOSIDES & NUCLEOTIDES, vol. 14, 1995, pages 969 - 973
MANOHARAN ET AL., TETRAHEDRON LETT., vol. 36, 1995, pages 3651 - 3654
MISHRA ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 1264, 1995, pages 229 - 237
MOL. ENDOCRINOL., vol. 21, 2007, pages 1769
MOL. PSYCHIATRY, vol. 11, 2006, pages 76
NAGAHAMA K ET AL: "Nuclease resistant methylphosphonate-DNA/LNA chimeric oligonucleotides", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 19, no. 10, 15 May 2009 (2009-05-15), pages 2707 - 2709, XP026085948, ISSN: 0960-894X, [retrieved on 20090328], DOI: 10.1016/J.BMCL.2009.03.116 *
NAT. GENET., vol. 33, 2003, pages 487
NAT. MED., vol. 3, 1997, pages 1009
NEUROBIOL DIS, vol. 3, 1996, pages 183
NEW ENGL J MED, vol. 346, 2002, pages 45
OBERHAUSER ET AL., NUCL. ACIDS RES., vol. 20, 1992, pages 533 - 538
ORUM ET AL., CURR. OPINION MOL. THER., vol. 3, 2001, pages 239 - 243
OWEN ET AL., J. ORG. CHEM., vol. 41, 1976, pages 3010 - 3017
PFISTER E L ET AL: "Five siRNAs Targeting Three SNPs May Provide Therapy for Three-Quarters of Huntington's Disease Patients", CURRENT BIOLOGY, CURRENT SCIENCE, GB, vol. 19, no. 9, 12 May 2009 (2009-05-12), pages 774 - 778, XP026099659, ISSN: 0960-9822, [retrieved on 20090409], DOI: 10.1016/J.CUB.2009.03.030 *
PLOS ONE, vol. 3, 2008, pages e3341
PLOS ONE, vol. 4, 2009, pages e7232
PROC. NATL. ASSOC. SCI., vol. 104, 2007, pages 19767
PROC. NATL. ASSOC. SCI., vol. 105, 2008, pages 4533
PROTEIN SCI, vol. 12, 2003, pages 953
SAISON-BEHMOARAS ET AL., EMBO J., vol. 10, 1991, pages 1111 - 1118
SCIENCE, vol. 281, 1998, pages 1851
SCIENCE, vol. 312, 2006, pages 1215
SHEA ET AL., NUCL. ACIDS RES., vol. 18, 1990, pages 3777 - 3783
SINGH ET AL., CHEM. COMMUN., 4 April 1998 (1998-04-04)
SINGH ET AL., J. ORG. CHEM., vol. 63, 1998, pages 10035 - 10039
SRIVASTAVA ET AL., J. AM. CHEM. SOC., vol. 129, no. 26, 2007, pages 8362 - 8379
SRIVASTAVA ET AL., J. AM. CHEM. SOC., vol. 129, no. 26, 4 July 2007 (2007-07-04), pages 8362 - 8379
SVINARCHUK ET AL., BIOCHIMIE, vol. 75, 1993, pages 49 - 54
TANG ET AL., J. ORG. CHEM., vol. 64, 1999, pages 747 - 754
TRANSL. RES., vol. 149, 2007, pages 205
TRENDS MOL. MED., vol. 7, 2001, pages 479
TRENDS PHARMACOL. SCI., vol. 27, 2006, pages 260
WAHLESTEDT ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 97, 2000, pages 5633 - 5638

Also Published As

Publication number Publication date
ES2635866T3 (es) 2017-10-05
US11732261B2 (en) 2023-08-22
EP2742056A4 (fr) 2015-03-25
WO2013022966A1 (fr) 2013-02-14
EP2742136A1 (fr) 2014-06-18
US20150051389A1 (en) 2015-02-19
EP2742056B2 (fr) 2020-06-10
EP2742056A1 (fr) 2014-06-18
EP3556859A1 (fr) 2019-10-23
EP3556859B1 (fr) 2021-04-07
EP2742136B1 (fr) 2017-09-27
US20230113863A1 (en) 2023-04-13
ES2635866T5 (es) 2021-04-05
WO2013022984A1 (fr) 2013-02-14
ES2651514T3 (es) 2018-01-26
EP2742135B2 (fr) 2020-06-10
US20210238591A1 (en) 2021-08-05
EP4269584A2 (fr) 2023-11-01
EP3205725A1 (fr) 2017-08-16
EP4269584A3 (fr) 2024-03-27
US20190338281A1 (en) 2019-11-07
DK2742135T4 (da) 2020-07-13
US20140323707A1 (en) 2014-10-30
EP2742135A1 (fr) 2014-06-18
EP3205725B1 (fr) 2019-03-27
US20140309279A1 (en) 2014-10-16
US9752142B2 (en) 2017-09-05
WO2013022967A1 (fr) 2013-02-14
WO2013022990A1 (fr) 2013-02-14
US20140303235A1 (en) 2014-10-09
EP2742135B1 (fr) 2017-05-03
DK2742135T3 (en) 2017-07-10
US20170130224A1 (en) 2017-05-11
DK2742136T3 (da) 2017-11-20
US20210147838A1 (en) 2021-05-20
US10202599B2 (en) 2019-02-12
EP3922722B1 (fr) 2023-06-28
EP2742056B1 (fr) 2017-01-11

Similar Documents

Publication Publication Date Title
US11732261B2 (en) Selective antisense compounds and uses thereof
US11236335B2 (en) Selective antisense compounds and uses thereof
US11492615B2 (en) Antisense compounds and uses thereof
EP2951304B1 (fr) Composés antisens sélectifs et leurs utilisations
EP3194628A1 (fr) Composés antisens et leurs utilisations

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2742056

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3205725

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3556859

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

B565 Issuance of search results under rule 164(2) epc

Effective date: 20211112

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20220609

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20230201

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AC Divisional application: reference to earlier application

Ref document number: 2742056

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3205725

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3556859

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230521

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1582737

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230715

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602012079811

Country of ref document: DE

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230928

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230629

Year of fee payment: 12

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20230628

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1582737

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230929

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230730

Year of fee payment: 12

Ref country code: DE

Payment date: 20230620

Year of fee payment: 12

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231030

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231028

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230808